1
|
Sherlock BD, Boon MAA, Vlasiou M, Coster ACF. The Distance Between: An Algorithmic Approach to Comparing Stochastic Models to Time-Series Data. Bull Math Biol 2024; 86:111. [PMID: 39060776 PMCID: PMC11282162 DOI: 10.1007/s11538-024-01331-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/21/2024] [Indexed: 07/28/2024]
Abstract
While mean-field models of cellular operations have identified dominant processes at the macroscopic scale, stochastic models may provide further insight into mechanisms at the molecular scale. In order to identify plausible stochastic models, quantitative comparisons between the models and the experimental data are required. The data for these systems have small sample sizes and time-evolving distributions. The aim of this study is to identify appropriate distance metrics for the quantitative comparison of stochastic model outputs and time-evolving stochastic measurements of a system. We identify distance metrics with features suitable for driving parameter inference, model comparison, and model validation, constrained by data from multiple experimental protocols. In this study, stochastic model outputs are compared to synthetic data across three scales: that of the data at the points the system is sampled during the time course of each type of experiment; a combined distance across the time course of each experiment; and a combined distance across all the experiments. Two broad categories of comparators at each point were considered, based on the empirical cumulative distribution function (ECDF) of the data and of the model outputs: discrete based measures such as the Kolmogorov-Smirnov distance, and integrated measures such as the Wasserstein-1 distance between the ECDFs. It was found that the discrete based measures were highly sensitive to parameter changes near the synthetic data parameters, but were largely insensitive otherwise, whereas the integrated distances had smoother transitions as the parameters approached the true values. The integrated measures were also found to be robust to noise added to the synthetic data, replicating experimental error. The characteristics of the identified distances provides the basis for the design of an algorithm suitable for fitting stochastic models to real world stochastic data.
Collapse
Affiliation(s)
- Brock D Sherlock
- School of Mathematics and Statistics, University of New South Wales, Sydney, NSW, 2052, Australia
- Department of Mathematics and Computer Science, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, The Netherlands
| | - Marko A A Boon
- Department of Mathematics and Computer Science, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, The Netherlands
| | - Maria Vlasiou
- Faculty of Electrical Engineering, Mathematics and Computer Science, University of Twente, P.O. Box 217, 7500 AE, Enschede, The Netherlands
| | - Adelle C F Coster
- School of Mathematics and Statistics, University of New South Wales, Sydney, NSW, 2052, Australia.
| |
Collapse
|
2
|
Ray A, Wen J, Yammine L, Culver J, Parida IS, Garren J, Xue L, Hales K, Xiang Q, Birnbaum MJ, Zhang BB, Monetti M, McGraw TE. Regulated dynamic subcellular GLUT4 localization revealed by proximal proteome mapping in human muscle cells. J Cell Sci 2023; 136:jcs261454. [PMID: 38126809 PMCID: PMC10753500 DOI: 10.1242/jcs.261454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
Regulation of glucose transport, which is central for control of whole-body metabolism, is determined by the amount of GLUT4 glucose transporter (also known as SLC2A4) in the plasma membrane (PM) of fat and muscle cells. Physiologic signals [such as activated insulin receptor or AMP-activated protein kinase (AMPK)] increase PM GLUT4. Here, we show that the distribution of GLUT4 between the PM and interior of human muscle cells is dynamically maintained, and that AMPK promotes PM redistribution of GLUT4 by regulating exocytosis and endocytosis. Stimulation of exocytosis by AMPK is mediated by Rab10 and the Rab GTPase-activating protein TBC1D4. APEX2 proximity mapping reveals that GLUT4 traverses both PM-proximal and PM-distal compartments in unstimulated muscle cells, further supporting retention of GLUT4 by a constitutive retrieval mechanism. AMPK-stimulated translocation involves GLUT4 redistribution among the same compartments traversed in unstimulated cells, with a significant recruitment of GLUT4 from the Golgi and trans-Golgi network compartments. Our comprehensive proximal protein mapping provides an integrated, high-density, whole-cell accounting of the localization of GLUT4 at a resolution of ∼20 nm that serves as a structural framework for understanding the molecular mechanisms regulating GLUT4 trafficking downstream of different signaling inputs in a physiologically relevant cell type.
Collapse
Affiliation(s)
- Anuttoma Ray
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jennifer Wen
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10021, USA
| | - Lucie Yammine
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jeff Culver
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development and Medical, Cambridge, MA 02139, USA
| | | | - Jeonifer Garren
- Global Biometrics and Data Management, Global Product Development, Pfizer Inc., Cambridge, MA 02139, USA
| | - Liang Xue
- Early Clinical Development Biomedicine AI, Pfizer Worldwide Research, Development and Medical, Cambridge, MA 02139, USA
| | - Katherine Hales
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development and Medical, Cambridge, MA 02139, USA
| | - Qing Xiang
- Target Sciences, Pfizer Inc., New York, NY 10016, USA
| | - Morris J. Birnbaum
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development and Medical, Cambridge, MA 02139, USA
| | - Bei B. Zhang
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development and Medical, Cambridge, MA 02139, USA
| | - Mara Monetti
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development and Medical, Cambridge, MA 02139, USA
| | - Timothy E. McGraw
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
3
|
Ray A, Wen J, Yammine L, Culver J, Garren J, Xue L, Hales K, Xiang Q, Birnbaum MJ, Zhang BB, Monetti M, McGraw TE. GLUT4 dynamic subcellular localization is controlled by AMP kinase activation as revealed by proximal proteome mapping in human muscle cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.543897. [PMID: 37333333 PMCID: PMC10274730 DOI: 10.1101/2023.06.06.543897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Regulation of glucose transport into muscle and adipocytes, central for control of whole-body metabolism, is determined by the amount of GLUT4 glucose transporter in the plasma membrane ( PM ). Physiologic signals (activated insulin receptor or AMP kinase [ AMPK ]), acutely increase PM GLUT4 to enhance glucose uptake. Here we show in kinetic studies that intracellular GLUT4 is in equilibrium with the PM in unstimulated cultured human skeletal muscle cells, and that AMPK promotes GLUT4 redistribution to the PM by regulating both exocytosis and endocytosis. AMPK-stimulation of exocytosis requires Rab10 and Rab GTPase activating protein TBC1D4, requirements shared with insulin control of GLUT4 in adipocytes. Using APEX2 proximity mapping, we identify, at high-density and high-resolution, the GLUT4 proximal proteome, revealing GLUT4 traverses both PM proximal and distal compartments in unstimulated muscle cells. These data support intracellular retention of GLUT4 in unstimulated muscle cells by a dynamic mechanism dependent on the rates of internalization and recycling. AMPK promoted GLUT4 translocation to the PM involves redistribution of GLUT4 among the same compartments traversed in unstimulated cells, with a significant redistribution of GLUT4 from the PM distal Trans Golgi Network Golgi compartments. The comprehensive proximal protein mapping provides an integrated, whole cell accounting of GLUT4's localization at a resolution of ∼20 nm, a structural framework for understanding the molecular mechanisms regulating GLUT4 trafficking downstream of different signaling inputs in physiologically relevant cell type and as such, sheds new light on novel key pathways and molecular components as potential therapeutic approaches to modulate muscle glucose uptake.
Collapse
|
4
|
Dennis KMJH, Heather LC. Post-translational palmitoylation of metabolic proteins. Front Physiol 2023; 14:1122895. [PMID: 36909239 PMCID: PMC9998952 DOI: 10.3389/fphys.2023.1122895] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/03/2023] [Indexed: 03/14/2023] Open
Abstract
Numerous cellular proteins are post-translationally modified by addition of a lipid group to their structure, which dynamically influences the proteome by increasing hydrophobicity of proteins often impacting protein conformation, localization, stability, and binding affinity. These lipid modifications include myristoylation and palmitoylation. Palmitoylation involves a 16-carbon saturated fatty acyl chain being covalently linked to a cysteine thiol through a thioester bond. Palmitoylation is unique within this group of modifications, as the addition of the palmitoyl group is reversible and enzyme driven, rapidly affecting protein targeting, stability and subcellular trafficking. The palmitoylation reaction is catalyzed by a large family of Asp-His-His-Cys (DHHCs) motif-containing palmitoyl acyltransferases, while the reverse reaction is catalyzed by acyl-protein thioesterases (APTs), that remove the acyl chain. Palmitoyl-CoA serves an important dual purpose as it is not only a key metabolite fueling energy metabolism, but is also a substrate for this PTM. In this review, we discuss protein palmitoylation in regulating substrate metabolism, focusing on membrane transport proteins and kinases that participate in substrate uptake into the cell. We then explore the palmitoylation of mitochondrial proteins and the palmitoylation regulatory enzymes, a less explored field for potential lipid metabolic regulation.
Collapse
Affiliation(s)
- Kaitlyn M J H Dennis
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Lisa C Heather
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
5
|
Fazakerley DJ, Koumanov F, Holman GD. GLUT4 On the move. Biochem J 2022; 479:445-462. [PMID: 35147164 PMCID: PMC8883492 DOI: 10.1042/bcj20210073] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 12/16/2022]
Abstract
Insulin rapidly stimulates GLUT4 translocation and glucose transport in fat and muscle cells. Signals from the occupied insulin receptor are translated into downstream signalling changes in serine/threonine kinases within timescales of seconds, and this is followed by delivery and accumulation of the glucose transporter GLUT4 at the plasma membrane. Kinetic studies have led to realisation that there are distinct phases of this stimulation by insulin. There is a rapid initial burst of GLUT4 delivered to the cell surface from a subcellular reservoir compartment and this is followed by a steady-state level of continuing stimulation in which GLUT4 recycles through a large itinerary of subcellular locations. Here, we provide an overview of the phases of insulin stimulation of GLUT4 translocation and the molecules that are currently considered to activate these trafficking steps. Furthermore, we suggest how use of new experimental approaches together with phospho-proteomic data may help to further identify mechanisms for activation of these trafficking processes.
Collapse
Affiliation(s)
- Daniel J Fazakerley
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, U.K
| | - Francoise Koumanov
- Department for Health, Centre for Nutrition, Exercise, and Metabolism, University of Bath, Bath, Somerset BA2 7AY, U.K
| | - Geoffrey D Holman
- Department of Biology and Biochemistry, University of Bath, Bath, Somerset BA2 7AY, U.K
| |
Collapse
|
6
|
Bogan JS. Ubiquitin-like processing of TUG proteins as a mechanism to regulate glucose uptake and energy metabolism in fat and muscle. Front Endocrinol (Lausanne) 2022; 13:1019405. [PMID: 36246906 PMCID: PMC9556833 DOI: 10.3389/fendo.2022.1019405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/06/2022] [Indexed: 12/02/2022] Open
Abstract
In response to insulin stimulation, fat and muscle cells mobilize GLUT4 glucose transporters to the cell surface to enhance glucose uptake. Ubiquitin-like processing of TUG (Aspscr1, UBXD9) proteins is a central mechanism to regulate this process. Here, recent advances in this area are reviewed. The data support a model in which intact TUG traps insulin-responsive "GLUT4 storage vesicles" at the Golgi matrix by binding vesicle cargoes with its N-terminus and matrix proteins with its C-terminus. Insulin stimulation liberates these vesicles by triggering endoproteolytic cleavage of TUG, mediated by the Usp25m protease. Cleavage occurs in fat and muscle cells, but not in fibroblasts or other cell types. Proteolytic processing of intact TUG generates TUGUL, a ubiquitin-like protein modifier, as the N-terminal cleavage product. In adipocytes, TUGUL modifies a single protein, the KIF5B kinesin motor, which carries GLUT4 and other vesicle cargoes to the cell surface. In muscle, this or another motor may be modified. After cleavage of intact TUG, the TUG C-terminal product is extracted from the Golgi matrix by the p97 (VCP) ATPase. In both muscle and fat, this cleavage product enters the nucleus, binds PPARγ and PGC-1α, and regulates gene expression to promote fatty acid oxidation and thermogenesis. The stability of the TUG C-terminal product is regulated by an Ate1 arginyltransferase-dependent N-degron pathway, which may create a feedback mechanism to control oxidative metabolism. Although it is now clear that TUG processing coordinates glucose uptake with other aspects of physiology and metabolism, many questions remain about how this pathway is regulated and how it is altered in metabolic disease in humans.
Collapse
Affiliation(s)
- Jonathan S. Bogan
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, United States
- Yale Center for Molecular and Systems Metabolism, Yale School of Medicine, New Haven, CT, United States
- *Correspondence: Jonathan S. Bogan,
| |
Collapse
|
7
|
Batty SR, Langlais PR. Microtubules in insulin action: what's on the tube? Trends Endocrinol Metab 2021; 32:776-789. [PMID: 34462181 PMCID: PMC8446328 DOI: 10.1016/j.tem.2021.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 11/17/2022]
Abstract
Microtubules (MT) have a role in the intracellular response to insulin stimulation and subsequent glucose transport by glucose transporter 4 (GLUT4), which resides in specialized storage vesicles that travel through the cell. Before GLUT4 is inserted into the plasma membrane for glucose transport, it undergoes complex trafficking through the cell via the integration of cytoskeletal networks. In this review, we highlight the importance of MT elements in insulin action in adipocytes through a summary of MT depolymerization studies, MT-based GLUT4 movement, molecular motor proteins involved in GLUT4 trafficking, as well as MT-related phenomena in response to insulin and links between insulin action and MT-associated proteins.
Collapse
Affiliation(s)
- Skylar R Batty
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, USA
| | - Paul R Langlais
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson, AZ, USA.
| |
Collapse
|
8
|
The Role of Overexpressed Apolipoprotein AV in Insulin-Resistant Hepatocytes. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3268505. [PMID: 32382544 PMCID: PMC7193279 DOI: 10.1155/2020/3268505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 07/16/2019] [Accepted: 08/16/2019] [Indexed: 11/17/2022]
Abstract
In this paper, we sought to explore the relationship between apolipoprotein AV (APOAV) overexpression and insulin resistance in hepatocytes. The insulin-resistant HepG2 cell model was constructed, and then, APOAV-overexpressed HepG2 cells (B-M) were induced by infecting with a recombinant adenovirus vector. Microarray data were developed from B-M samples compared with negative controls (A-con), and the microarray data were analyzed by bioinformatic methods. APOAV-overexpression induced 313 upregulated genes and 563 downregulated ones in B-M sample. The differentially expressed genes (DEGs) were significantly classified in fat digestion and absorption pathway. Protein-protein interaction network was constructed, and AGTR1 (angiotensin II receptor type 1) and P2RY2 (purinergic receptor P2Y, G-protein coupled 2) were found to be the significant nodes closely related with G-protein related signaling. Additionally, overexpression of APOAV could change the expression of Glut4 and release the insulin resistance of hepatic cells. Thus, APOAV overexpression may prevent the insulin resistance in liver cells by mediating the genes such as AGTR1 and P2RY2.
Collapse
|
9
|
Morris S, Geoghegan ND, Sadler JBA, Koester AM, Black HL, Laub M, Miller L, Heffernan L, Simpson JC, Mastick CC, Cooper J, Gadegaard N, Bryant NJ, Gould GW. Characterisation of GLUT4 trafficking in HeLa cells: comparable kinetics and orthologous trafficking mechanisms to 3T3-L1 adipocytes. PeerJ 2020; 8:e8751. [PMID: 32185116 PMCID: PMC7060922 DOI: 10.7717/peerj.8751] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/14/2020] [Indexed: 12/18/2022] Open
Abstract
Insulin-stimulated glucose transport is a characteristic property of adipocytes and muscle cells and involves the regulated delivery of glucose transporter (GLUT4)-containing vesicles from intracellular stores to the cell surface. Fusion of these vesicles results in increased numbers of GLUT4 molecules at the cell surface. In an attempt to overcome some of the limitations associated with both primary and cultured adipocytes, we expressed an epitope- and GFP-tagged version of GLUT4 (HA–GLUT4–GFP) in HeLa cells. Here we report the characterisation of this system compared to 3T3-L1 adipocytes. We show that insulin promotes translocation of HA–GLUT4–GFP to the surface of both cell types with similar kinetics using orthologous trafficking machinery. While the magnitude of the insulin-stimulated translocation of GLUT4 is smaller than mouse 3T3-L1 adipocytes, HeLa cells offer a useful, experimentally tractable, human model system. Here, we exemplify their utility through a small-scale siRNA screen to identify GOSR1 and YKT6 as potential novel regulators of GLUT4 trafficking in human cells.
Collapse
Affiliation(s)
- Silke Morris
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, UK
| | | | - Jessica B A Sadler
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, UK
| | - Anna M Koester
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, UK
| | | | - Marco Laub
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, UK
| | - Lucy Miller
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, UK
| | - Linda Heffernan
- School of Biology & Environmental Science, University College Dublin, Dublin, Ireland
| | - Jeremy C Simpson
- School of Biology & Environmental Science, University College Dublin, Dublin, Ireland
| | | | - Jon Cooper
- School of Engineering, University of Glasgow, Glasgow, UK
| | | | - Nia J Bryant
- Department of Biology, University of York, York, UK
| | - Gwyn W Gould
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
10
|
Li DT, Habtemichael EN, Julca O, Sales CI, Westergaard XO, DeVries SG, Ruiz D, Sayal B, Bogan JS. GLUT4 Storage Vesicles: Specialized Organelles for Regulated Trafficking. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2019; 92:453-470. [PMID: 31543708 PMCID: PMC6747935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Fat and muscle cells contain a specialized, intracellular organelle known as the GLUT4 storage vesicle (GSV). Insulin stimulation mobilizes GSVs, so that these vesicles fuse at the cell surface and insert GLUT4 glucose transporters into the plasma membrane. This example is likely one instance of a broader paradigm for regulated, non-secretory exocytosis, in which intracellular vesicles are translocated in response to diverse extracellular stimuli. GSVs have been studied extensively, yet these vesicles remain enigmatic. Data support the view that in unstimulated cells, GSVs are present as a pool of preformed small vesicles, which are distinct from endosomes and other membrane-bound organelles. In adipocytes, GSVs contain specific cargoes including GLUT4, IRAP, LRP1, and sortilin. They are formed by membrane budding, involving sortilin and probably CHC22 clathrin in humans, but the donor compartment from which these vesicles form remains uncertain. In unstimulated cells, GSVs are trapped by TUG proteins near the endoplasmic reticulum - Golgi intermediate compartment (ERGIC). Insulin signals through two main pathways to mobilize these vesicles. Signaling by the Akt kinase modulates Rab GTPases to target the GSVs to the cell surface. Signaling by the Rho-family GTPase TC10α stimulates Usp25m-mediated TUG cleavage to liberate the vesicles from the Golgi. Cleavage produces a ubiquitin-like protein modifier, TUGUL, that links the GSVs to KIF5B kinesin motors to promote their movement to the cell surface. In obesity, attenuation of these processes results in insulin resistance and contributes to type 2 diabetes and may simultaneously contribute to hypertension and dyslipidemia in the metabolic syndrome.
Collapse
Affiliation(s)
- Don T. Li
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, Yale University, New Haven, CT,Department of Cell Biology, Yale University School of Medicine, Yale University, New Haven, CT
| | - Estifanos N. Habtemichael
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, Yale University, New Haven, CT
| | - Omar Julca
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, Yale University, New Haven, CT
| | - Chloe I. Sales
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, Yale University, New Haven, CT
| | - Xavier O. Westergaard
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, Yale University, New Haven, CT
| | - Stephen G. DeVries
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, Yale University, New Haven, CT
| | - Diana Ruiz
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, Yale University, New Haven, CT
| | - Bhavesh Sayal
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, Yale University, New Haven, CT
| | - Jonathan S. Bogan
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, Yale University, New Haven, CT,Department of Cell Biology, Yale University School of Medicine, Yale University, New Haven, CT,To whom all correspondence should be addressed: Jonathan S. Bogan, Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, P.O. Box 208020, New Haven, CT 06520-8020; Tel: 203-785-6319; Fax: 203-785-6462;
| |
Collapse
|
11
|
Sphingolipid Metabolism: New Insight into Ceramide-Induced Lipotoxicity in Muscle Cells. Int J Mol Sci 2019; 20:ijms20030479. [PMID: 30678043 PMCID: PMC6387241 DOI: 10.3390/ijms20030479] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/10/2019] [Accepted: 01/18/2019] [Indexed: 12/17/2022] Open
Abstract
Insulin-resistance is a characteristic feature of type 2 diabetes (T2D) and plays a major role in the pathogenesis of this disease. Skeletal muscles are quantitatively the biggest glucose users in response to insulin and are considered as main targets in development of insulin-resistance. It is now clear that circulating fatty acids (FA), which are highly increased in T2D, play a major role in the development of muscle insulin-resistance. In healthy individuals, excess FA are stored as lipid droplets in adipocytes. In situations like obesity and T2D, FA from lipolysis and food are in excess and eventually accumulate in peripheral tissues. High plasma concentrations of FA are generally associated with increased risk of developing diabetes. Indeed, ectopic fat accumulation is associated with insulin-resistance; this is called lipotoxicity. However, FA themselves are not involved in insulin-resistance, but rather some of their metabolic derivatives, such as ceramides. Ceramides, which are synthetized de novo from saturated FA like palmitate, have been demonstrated to play a critical role in the deterioration of insulin sensitivity in muscle cells. This review describes the latest progress involving ceramides as major players in the development of muscle insulin-resistance through the targeting of selective actors of the insulin signaling pathway.
Collapse
|
12
|
Tokarz VL, MacDonald PE, Klip A. The cell biology of systemic insulin function. J Cell Biol 2018; 217:2273-2289. [PMID: 29622564 PMCID: PMC6028526 DOI: 10.1083/jcb.201802095] [Citation(s) in RCA: 235] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/21/2018] [Accepted: 03/23/2018] [Indexed: 12/12/2022] Open
Abstract
Insulin is the paramount anabolic hormone, promoting carbon energy deposition in the body. Its synthesis, quality control, delivery, and action are exquisitely regulated by highly orchestrated intracellular mechanisms in different organs or "stations" of its bodily journey. In this Beyond the Cell review, we focus on these five stages of the journey of insulin through the body and the captivating cell biology that underlies the interaction of insulin with each organ. We first analyze insulin's biosynthesis in and export from the β-cells of the pancreas. Next, we focus on its first pass and partial clearance in the liver with its temporality and periodicity linked to secretion. Continuing the journey, we briefly describe insulin's action on the blood vasculature and its still-debated mechanisms of exit from the capillary beds. Once in the parenchymal interstitium of muscle and adipose tissue, insulin promotes glucose uptake into myofibers and adipocytes, and we elaborate on the intricate signaling and vesicle traffic mechanisms that underlie this fundamental function. Finally, we touch upon the renal degradation of insulin to end its action. Cellular discernment of insulin's availability and action should prove critical to understanding its pivotal physiological functions and how their failure leads to diabetes.
Collapse
Affiliation(s)
- Victoria L Tokarz
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Patrick E MacDonald
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
13
|
Norris DM, Geddes TA, James DE, Fazakerley DJ, Burchfield JG. Glucose Transport: Methods for Interrogating GLUT4 Trafficking in Adipocytes. Methods Mol Biol 2018; 1713:193-215. [PMID: 29218527 DOI: 10.1007/978-1-4939-7507-5_15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In this chapter we detail methods for the systematic dissection of GLUT4 trafficking. The methods described have been optimized for cultured 3T3-L1 adipocytes, but can be readily adapted to other cell types.
Collapse
Affiliation(s)
- Dougall M Norris
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
- The Charles Perkins Centre, The University of Sydney, Sydney, 2006, NSW, Australia
| | - Tom A Geddes
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
- The Charles Perkins Centre, The University of Sydney, Sydney, 2006, NSW, Australia
| | - David E James
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, 2006, Australia
- The Charles Perkins Centre, The University of Sydney, Sydney, 2006, NSW, Australia
| | - Daniel J Fazakerley
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
- The Charles Perkins Centre, The University of Sydney, Sydney, 2006, NSW, Australia
| | - James G Burchfield
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia.
- The Charles Perkins Centre, The University of Sydney, Sydney, 2006, NSW, Australia.
| |
Collapse
|
14
|
Rab14 limits the sorting of Glut4 from endosomes into insulin-sensitive regulated secretory compartments in adipocytes. Biochem J 2016; 473:1315-27. [PMID: 26936971 DOI: 10.1042/bcj20160020] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 03/01/2016] [Indexed: 12/31/2022]
Abstract
Insulin increases glucose uptake by increasing the rate of exocytosis of the facilitative glucose transporter isoform 4 (Glut4) relative to its endocytosis. Insulin also releases Glut4 from highly insulin-regulated secretory compartments (GSVs or Glut4 storage vesicles) into constitutively cycling endosomes. Previously it was shown that both overexpression and knockdown of the small GTP-binding protein Rab14 decreased Glut4 translocation to the plasma membrane (PM). To determine the mechanism of this perturbation, we measured the effects of Rab14 knockdown on the trafficking kinetics of Glut4 relative to two proteins that partially co-localize with Glut4, the transferrin (Tf) receptor and low-density-lipoprotein-receptor-related protein 1 (LRP1). Our data support the hypothesis that Rab14 limits sorting of proteins from sorting (or 'early') endosomes into the specialized GSV pathway, possibly through regulation of endosomal maturation. This hypothesis is consistent with known Rab14 effectors. Interestingly, the insulin-sensitive Rab GTPase-activating protein Akt substrate of 160 kDa (AS160) affects both sorting into and exocytosis from GSVs. It has previously been shown that exocytosis of GSVs is rate-limited by Rab10, and both Rab10 and Rab14 are in vitro substrates of AS160. Regulation of both entry into and exit from GSVs by AS160 through sequential Rab substrates would provide a mechanism for the finely tuned 'quantal' increases in cycling Glut4 observed in response to increasing concentrations of insulin.
Collapse
|
15
|
Brewer PD, Habtemichael EN, Romenskaia I, Mastick CC, Coster ACF. Glut4 Is Sorted from a Rab10 GTPase-independent Constitutive Recycling Pathway into a Highly Insulin-responsive Rab10 GTPase-dependent Sequestration Pathway after Adipocyte Differentiation. J Biol Chem 2015; 291:773-89. [PMID: 26527681 DOI: 10.1074/jbc.m115.694919] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Indexed: 02/04/2023] Open
Abstract
The RabGAP AS160/TBC1D4 controls exocytosis of the insulin-sensitive glucose transporter Glut4 in adipocytes. Glut4 is internalized and recycled through a highly regulated secretory pathway in these cells. Glut4 also cycles through a slow constitutive endosomal pathway distinct from the fast transferrin (Tf) receptor recycling pathway. This slow constitutive pathway is the only Glut4 cycling pathway in undifferentiated fibroblasts. The α2-macroglobulin receptor LRP1 cycles with Glut4 and the Tf receptor through all three exocytic pathways. To further characterize these pathways, the effects of knockdown of AS160 substrates on the trafficking kinetics of Glut4, LRP1, and the Tf receptor were measured in adipocytes and fibroblasts. Rab10 knockdown decreased cell surface Glut4 in insulin-stimulated adipocytes by 65%, but not in basal adipocytes or in fibroblasts. This decrease was due primarily to a 62% decrease in the rate constant of Glut4 exocytosis (kex), although Rab10 knockdown also caused a 1.4-fold increase in the rate constant of Glut4 endocytosis (ken). Rab10 knockdown in adipocytes also decreased cell surface LRP1 by 30% by decreasing kex 30-40%. There was no effect on LRP1 trafficking in fibroblasts or on Tf receptor trafficking in either cell type. These data confirm that Rab10 is an AS160 substrate that limits exocytosis through the highly insulin-responsive specialized secretory pathway in adipocytes. They further show that the slow constitutive endosomal (fibroblast) recycling pathway is Rab10-independent. Thus, Rab10 is a marker for the specialized pathway in adipocytes. Interestingly, mathematical modeling shows that Glut4 traffics predominantly through the specialized Rab10-dependent pathway both before and after insulin stimulation.
Collapse
Affiliation(s)
- Paul Duffield Brewer
- From the Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Nevada 89557
| | - Estifanos N Habtemichael
- the Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06520, and
| | - Irina Romenskaia
- From the Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Nevada 89557
| | - Cynthia Corley Mastick
- From the Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Nevada 89557,
| | - Adelle C F Coster
- the Department of Applied Mathematics, School of Mathematics and Statistics, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
16
|
Alves DS, Thulin G, Loffing J, Kashgarian M, Caplan MJ. Akt Substrate of 160 kD Regulates Na+,K+-ATPase Trafficking in Response to Energy Depletion and Renal Ischemia. J Am Soc Nephrol 2015; 26:2765-76. [PMID: 25788531 DOI: 10.1681/asn.2013101040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 01/06/2015] [Indexed: 01/26/2023] Open
Abstract
Renal ischemia and reperfusion injury causes loss of renal epithelial cell polarity and perturbations in tubular solute and fluid transport. Na(+),K(+)-ATPase, which is normally found at the basolateral plasma membrane of renal epithelial cells, is internalized and accumulates in intracellular compartments after renal ischemic injury. We previously reported that the subcellular distribution of Na(+),K(+)-ATPase is modulated by direct binding to Akt substrate of 160 kD (AS160), a Rab GTPase-activating protein that regulates the trafficking of glucose transporter 4 in response to insulin and muscle contraction. Here, we investigated the effect of AS160 on Na(+),K(+)-ATPase trafficking in response to energy depletion. We found that AS160 is required for the intracellular accumulation of Na(+),K(+)-ATPase that occurs in response to energy depletion in cultured epithelial cells. Energy depletion led to dephosphorylation of AS160 at S588, which was required for the energy depletion-induced accumulation of Na,K-ATPase in intracellular compartments. In AS160-knockout mice, the effects of renal ischemia on the distribution of Na(+),K(+)-ATPase were substantially reduced in the epithelial cells of distal segments of the renal tubules. These data demonstrate that AS160 has a direct role in linking the trafficking of Na(+),K(+)-ATPase to the energy state of renal epithelial cells.
Collapse
Affiliation(s)
| | - Gunilla Thulin
- Pathology, Yale University School of Medicine, New Haven, Connecticut; and
| | | | - Michael Kashgarian
- Pathology, Yale University School of Medicine, New Haven, Connecticut; and
| | | |
Collapse
|
17
|
Govers R. Molecular mechanisms of GLUT4 regulation in adipocytes. DIABETES & METABOLISM 2014; 40:400-10. [DOI: 10.1016/j.diabet.2014.01.005] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 01/24/2014] [Accepted: 01/26/2014] [Indexed: 01/28/2023]
|
18
|
Tessneer KL, Jackson RM, Griesel BA, Olson AL. Rab5 activity regulates GLUT4 sorting into insulin-responsive and non-insulin-responsive endosomal compartments: a potential mechanism for development of insulin resistance. Endocrinology 2014; 155:3315-28. [PMID: 24932807 PMCID: PMC4138579 DOI: 10.1210/en.2013-2148] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Glucose transporter isoform 4 (GLUT4) is the insulin-responsive glucose transporter mediating glucose uptake in adipose and skeletal muscle. Reduced GLUT4 translocation from intracellular storage compartments to the plasma membrane is a cause of peripheral insulin resistance. Using a chronic hyperinsulinemia (CHI)-induced cell model of insulin resistance and Rab5 mutant overexpression, we determined these manipulations altered endosomal sorting of GLUT4, thus contributing to the development of insulin resistance. We found that CHI induced insulin resistance in 3T3-L1 adipocytes by retaining GLUT4 in a Rab5-activity-dependent compartment that is unable to equilibrate with the cell surface in response to insulin. Furthermore, CHI-mediated retention of GLUT4 in this non-insulin-responsive compartment impaired filling of the transferrin receptor (TfR)-positive and TfR-negative insulin-responsive storage compartments. Our data suggest that hyperinsulinemia may inhibit GLUT4 by chronically maintaining GLUT4 in the Rab5 activity-dependent endosomal pathway and impairing formation of the TfR-negative and TfR-positive insulin-responsive GLUT4 pools. This model suggests that an early event in the development of insulin-resistant glucose transport in adipose tissue is to alter the intracellular localization of GLUT4 to a compartment that does not efficiently equilibrate with the cell surface when insulin levels are elevated for prolonged periods of time.
Collapse
Affiliation(s)
- Kandice L Tessneer
- Department of Biochemistry and Molecular Biology (K.L.T., R.M.J., B.A.G., A.L.O.), University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73126; and Cardiovascular Biology Program (K.L.T.), Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| | | | | | | |
Collapse
|
19
|
Jezewski AJ, Larson JJ, Wysocki B, Davis PH, Wysocki T. A novel method for simulating insulin mediated GLUT4 translocation. Biotechnol Bioeng 2014; 111:2454-2465. [PMID: 24917169 DOI: 10.1002/bit.25310] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 04/22/2014] [Accepted: 06/05/2014] [Indexed: 01/19/2023]
Abstract
Glucose transport in humans is a vital process which is tightly regulated by the endocrine system. Specifically, the insulin hormone triggers a cascade of intracellular signals in target cells mediating the uptake of glucose. Insulin signaling triggers cellular relocalization of the glucose transporter protein GLUT4 to the cell surface, which is primarily responsible for regulated glucose import. Pathology associated with the disruption of this pathway can lead to metabolic disorders, such as type II diabetes mellitus, characterized by the failure of cells to appropriately uptake glucose from the blood. We describe a novel simulation tool of the insulin intracellular response, incorporating the latest findings regarding As160 and GEF interactions. The simulation tool differs from previous computational approaches which employ algebraic or differential equations; instead, the tool incorporates statistical variations of kinetic constants and initial molecular concentrations which more accurately mimic the intracellular environment. Using this approach, we successfully recapitulate observed in vitro insulin responses, plus the effects of Wortmannin-like inhibition of the pathway. The developed tool provides insight into transient changes in molecule concentrations throughout the insulin signaling pathway, and may be employed to identify or evaluate potentially critical components of this pathway, including those associated with insulin resistance. In the future, this highly tractable platform may be useful for simulating other complex cell signaling pathways. Biotechnol. Bioeng. 2014;111: 2454-2465. © 2014 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Andrew J Jezewski
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska
| | - Joshua J Larson
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska
| | - Beata Wysocki
- Department of Engineering, University of Nebraska-Lincoln, 6001 Dodge St, 200 Peter Kiewit Institute, Omaha, Nebraska 68182-0572;
| | - Paul H Davis
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska.,Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska
| | - Tadeusz Wysocki
- Department of Engineering, University of Nebraska-Lincoln, 6001 Dodge St, 200 Peter Kiewit Institute, Omaha, Nebraska 68182-0572;
| |
Collapse
|
20
|
Brewer PD, Habtemichael EN, Romenskaia I, Mastick CC, Coster ACF. Insulin-regulated Glut4 translocation: membrane protein trafficking with six distinctive steps. J Biol Chem 2014; 289:17280-98. [PMID: 24778187 DOI: 10.1074/jbc.m114.555714] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The trafficking kinetics of Glut4, the transferrin (Tf) receptor, and LRP1 were quantified in adipocytes and undifferentiated fibroblasts. Six steps were identified that determine steady state cell surface Glut4: (i) endocytosis, (ii) degradation, (iii) sorting, (iv) sequestration, (v) release, and (vi) tethering/docking/fusion. Endocytosis of Glut4 is 3 times slower than the Tf receptor in fibroblasts (ken = 0.2 min(-1) versus 0.6 min(-1)). Differentiation decreases Glut4 ken 40% (ken = 0.12 min(-1)). Differentiation also decreases Glut4 degradation, increasing total and cell surface Glut4 3-fold. In fibroblasts, Glut4 is recycled from endosomes through a slow constitutive pathway (kex = 0.025-0.038 min(-1)), not through the fast Tf receptor pathway (kex = 0.2 min(-1)). The kex measured in adipocytes after insulin stimulation is similar (kex = 0.027 min(-1)). Differentiation decreases the rate constant for sorting into the Glut4 recycling pathway (ksort) 3-fold. In adipocytes, Glut4 is also sorted from endosomes into a second exocytic pathway through Glut4 storage vesicles (GSVs). Surprisingly, transfer from endosomes into GSVs is highly regulated; insulin increases the rate constant for sequestration (kseq) 8-fold. Release from sequestration in GSVs is rate-limiting for Glut4 exocytosis in basal adipocytes. AS160 regulates this step. Tethering/docking/fusion of GSVs to the plasma membrane is regulated through an AS160-independent process. Insulin increases the rate of release and fusion of GSVs (kfuseG) 40-fold. LRP1 cycles with the Tf receptor and Glut4 in fibroblasts but predominantly with Glut4 after differentiation. Surprisingly, AS160 knockdown accelerated LRP1 exocytosis in basal and insulin-stimulated adipocytes. These data indicate that AS160 may regulate trafficking into as well as release from GSVs.
Collapse
Affiliation(s)
- Paul Duffield Brewer
- From the Department of Biochemistry and Molecular Biology, University of Nevada School of Medicine, Reno, Nevada 89557
| | - Estifanos N Habtemichael
- the Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06520, and
| | - Irina Romenskaia
- From the Department of Biochemistry and Molecular Biology, University of Nevada School of Medicine, Reno, Nevada 89557
| | - Cynthia Corley Mastick
- From the Department of Biochemistry and Molecular Biology, University of Nevada School of Medicine, Reno, Nevada 89557,
| | - Adelle C F Coster
- the School of Mathematics and Statistics, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
21
|
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
|
22
|
Song X, Lichti CF, Townsend RR, Mueckler M. Single point mutations result in the miss-sorting of Glut4 to a novel membrane compartment associated with stress granule proteins. PLoS One 2013; 8:e68516. [PMID: 23874650 PMCID: PMC3713040 DOI: 10.1371/journal.pone.0068516] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 05/29/2013] [Indexed: 01/16/2023] Open
Abstract
Insulin increases cellular glucose uptake and metabolism in the postprandial state by acutely stimulating the translocation of the Glut4 glucose transporter from intracellular membrane compartments to the cell surface in muscle and fat cells. The intracellular targeting of Glut4 is dictated by specific structural motifs within cytoplasmic domains of the transporter. We demonstrate that two leucine residues at the extreme C-terminus of Glut4 are critical components of a motif (IRM, insulin responsive motif) involved in the sorting of the transporter to insulin responsive vesicles in 3T3L1 adipocytes. Light microscopy, immunogold electron microscopy, subcellular fractionation, and sedimentation analysis indicate that mutations in the IRM cause the aberrant targeting of Glut4 to large dispersed membrane vesicles that are not insulin responsive. Proteomic characterization of rapidly and slowly sedimenting membrane vesicles (RSVs and SSVs) that were highly enriched by immunoadsorption for either wild-type Glut4 or an IRM mutant revealed that the major vesicle fraction containing the mutant transporter (IRM-RSVs) possessed a relatively small and highly distinct protein population that was enriched for proteins associated with stress granules. We suggest that the IRM is critical for an early step in the sorting of Glut4 to insulin-responsive subcellular membrane compartments and that IRM mutants are miss-targeted to relatively large, amorphous membrane vesicles that may be involved in a degradation pathway for miss-targeted or miss-folded proteins or represent a transitional membrane compartment that Glut4 traverses en route to insulin responsive storage compartments.
Collapse
Affiliation(s)
- XiaoMei Song
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Cheryl F. Lichti
- Department of Pharmacology & Toxicology, University of Texas, Galveston, Texas, United States of America
| | - R. Reid Townsend
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Mike Mueckler
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
23
|
Abstract
GLUT4 is an insulin-regulated glucose transporter that is responsible for insulin-regulated glucose uptake into fat and muscle cells. In the absence of insulin, GLUT4 is mainly found in intracellular vesicles referred to as GLUT4 storage vesicles (GSVs). Here, we summarise evidence for the existence of these specific vesicles, how they are sequestered inside the cell and how they undergo exocytosis in the presence of insulin. In response to insulin stimulation, GSVs fuse with the plasma membrane in a rapid burst and in the continued presence of insulin GLUT4 molecules are internalised and recycled back to the plasma membrane in vesicles that are distinct from GSVs and probably of endosomal origin. In this Commentary we discuss evidence that this delivery process is tightly regulated and involves numerous molecules. Key components include the actin cytoskeleton, myosin motors, several Rab GTPases, the exocyst, SNARE proteins and SNARE regulators. Each step in this process is carefully orchestrated in a sequential and coupled manner and we are beginning to dissect key nodes within this network that determine vesicle-membrane fusion in response to insulin. This regulatory process clearly involves the Ser/Thr kinase AKT and the exquisite manner in which this single metabolic process is regulated makes it a likely target for lesions that might contribute to metabolic disease.
Collapse
Affiliation(s)
- Jacqueline Stöckli
- Diabetes and Obesity Program, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
| | | | | |
Collapse
|
24
|
Abstract
To enhance glucose uptake into muscle and fat cells, insulin stimulates the translocation of GLUT4 glucose transporters from intracellular membranes to the cell surface. This response requires the intersection of insulin signaling and vesicle trafficking pathways, and it is compromised in the setting of overnutrition to cause insulin resistance. Insulin signals through AS160/Tbc1D4 and Tbc1D1 to modulate Rab GTPases and through the Rho GTPase TC10α to act on other targets. In unstimulated cells, GLUT4 is incorporated into specialized storage vesicles containing IRAP, LRP1, sortilin, and VAMP2, which are sequestered by TUG, Ubc9, and other proteins. Insulin mobilizes these vesicles directly to the plasma membrane, and it modulates the trafficking itinerary so that cargo recycles from endosomes during ongoing insulin exposure. Knowledge of how signaling and trafficking pathways are coordinated will be essential to understanding the pathogenesis of diabetes and the metabolic syndrome and may also inform a wide range of other physiologies.
Collapse
Affiliation(s)
- Jonathan S Bogan
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06520-8020, USA.
| |
Collapse
|
25
|
Hatakeyama H, Kanzaki M. Molecular basis of insulin-responsive GLUT4 trafficking systems revealed by single molecule imaging. Traffic 2011; 12:1805-20. [PMID: 21910807 DOI: 10.1111/j.1600-0854.2011.01279.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Development of a 'static retention' property of GLUT4, the insulin-responsive glucose transporter, has emerged as being essential for achieving its maximal insulin-induced surface exposure. Herein, employing quantum-dot-based nanometrology of intracellular GLUT4 behavior, we reveal the molecular basis of its systematization endowed upon adipogenic differentiation of 3T3L1 cells. Specifically, (i) the endosomes-to-trans-Golgi network (TGN) retrieval system specialized for GLUT4 develops in response to sortilin expression, which requires an intricately balanced interplay among retromers, golgin-97 and syntaxin-6, the housekeeping vesicle trafficking machinery. (ii) The Golgin-97-localizing subdomain of the differentiated TGN apparently serves as an intermediate transit route by which GLUT4 can further proceed to the stationary GLUT4 storage compartment. (iii) AS160/Tbc1d4 then renders the 'static retention' property insulin responsive, i.e. insulin liberates GLUT4 from the static state only in the presence of functional AS160/Tbc1d4. (iv) Moreover, sortilin malfunction and the resulting GLUT4 sorting defects along with retarded TGN function might be etiologically related to insulin resistance. Together, these observations provide a conceptual framework for understanding maturation/retardation of the insulin-responsive GLUT4 trafficking system that relies on the specialized subdomain of differentiated TGN.
Collapse
Affiliation(s)
- Hiroyasu Hatakeyama
- Graduate School of Biomedical Engineering, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | | |
Collapse
|
26
|
Stöckli J, Fazakerley DJ, Coster ACF, Holman GD, James DE. Muscling in on GLUT4 kinetics. Commun Integr Biol 2011; 3:260-2. [PMID: 20714409 DOI: 10.4161/cib.3.3.11457] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Accepted: 02/10/2010] [Indexed: 01/14/2023] Open
Abstract
Insulin triggers glucose uptake into muscle and adipose tissue by stimulating the translocation of the glucose transporter glut4 from intracellular vesicles to the plasma membrane (pm). insulin leads to a rapid increase in glut4 at the pm from approximately 5% to 40-50%. this effect is time and dose-dependent, reaching a new steady state after 30 min of insulin stimulation. previous kinetic analyses in adipocytes has revealed that this is regulated by two mechanisms-increasing the amount of glut4 in the endosomal recycling system and increasing the exocytosis rate constant. fazakerley et al.1 focuses on GLUT4 kinetics in the L6 skeletal muscle cell line. Despite displaying a similar redistribution of GLUT4 to the cell surface with insulin to that seen in adipocytes, the mechanism for this effect in L6 cells was completely different. Insulin had a modest effect to increase the amount of GLUT4 in the recycling system with the dominant effect being on reduction of the endocytosis rate constant. Similar findings were observed with AMPK agonists. These studies indicate that different cell types are capable of achieving the same cell biological endpoint but using completely distinct mechanisms.
Collapse
|
27
|
Brewer PD, Romenskaia I, Kanow MA, Mastick CC. Loss of AS160 Akt substrate causes Glut4 protein to accumulate in compartments that are primed for fusion in basal adipocytes. J Biol Chem 2011; 286:26287-97. [PMID: 21613213 DOI: 10.1074/jbc.m111.253880] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The Akt substrate AS160 (TCB1D4) regulates Glut4 exocytosis; shRNA knockdown of AS160 increases surface Glut4 in basal adipocytes. AS160 knockdown is only partially insulin-mimetic; insulin further stimulates Glut4 translocation in these cells. Insulin regulates translocation as follows: 1) by releasing Glut4 from retention in a slowly cycling/noncycling storage pool, increasing the actively cycling Glut4 pool, and 2) by increasing the intrinsic rate constant for exocytosis of the actively cycling pool (k(ex)). Kinetic studies were performed in 3T3-L1 adipocytes to measure the effects of AS160 knockdown on the rate constants of exocytosis (k(ex)), endocytosis (k(en)), and release from retention into the cycling pool. AS160 knockdown released Glut4 into the actively cycling pool without affecting k(ex) or k(en). Insulin increased k(ex) in the knockdown cells, further increasing cell surface Glut4. Inhibition of phosphatidylinositol 3-kinase or Akt affected both k(ex) and release from retention in control cells but only k(ex) in AS160 knockdown cells. Glut4 vesicles accumulate in a primed pre-fusion pool in basal AS160 knockdown cells. Akt regulates the rate of exocytosis of the primed vesicles through an AS160-independent mechanism. Therefore, there is an additional Akt substrate that regulates the fusion of Glut4 vesicles that remain to be identified. Mathematical modeling was used to test the hypothesis that this substrate regulates vesicle priming (release from retention), whereas AS160 regulates the reverse step by stimulating GTP turnover of a Rab protein required for vesicle tethering/docking/fusion. Our analysis indicates that fusion of the primed vesicles with the plasma membrane is an additional non-Akt-dependent insulin-regulated step.
Collapse
Affiliation(s)
- Paul Duffield Brewer
- Department of Biochemistry and Molecular Biology, University of Nevada School of Medicine, Reno, Nevada 89557, USA
| | | | | | | |
Collapse
|
28
|
Foley K, Boguslavsky S, Klip A. Endocytosis, recycling, and regulated exocytosis of glucose transporter 4. Biochemistry 2011; 50:3048-61. [PMID: 21405107 DOI: 10.1021/bi2000356] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Glucose transporter 4 (GLUT4) is responsible for the uptake of glucose into muscle and adipose tissues. Under resting conditions, GLUT4 is dynamically retained through idle cycling among selective intracellular compartments, from whence it undergoes slow recycling to the plasma membrane (PM). This dynamic retention can be released by command from intracellular signals elicited by insulin and other stimuli, which result in 2-10-fold increases in the surface level of GLUT4. Insulin-derived signals promote translocation of GLUT4 to the PM from a specialized compartment termed GLUT4 storage vesicles (GSV). Much effort has been devoted to the characterization of the intracellular compartments and dynamics of GLUT4 cycling and to the signals by which GLUT4 is sorted into, and recruited from, GSV. This review summarizes our understanding of intracellular GLUT4 traffic during its internalization from the membrane, its slow, constitutive recycling, and its regulated exocytosis in response to insulin. In spite of specific differences in GLUT4 dynamic behavior in adipose and muscle cells, the generalities of its endocytic and exocytic itineraries are consistent and an array of regulatory proteins that regulate each vesicular traffic event emerges from these cell systems.
Collapse
Affiliation(s)
- Kevin Foley
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario M4G 1X8, Canada
| | | | | |
Collapse
|
29
|
Habtemichael EN, Brewer PD, Romenskaia I, Mastick CC. Kinetic evidence that Glut4 follows different endocytic pathways than the receptors for transferrin and alpha2-macroglobulin. J Biol Chem 2011; 286:10115-25. [PMID: 21252237 DOI: 10.1074/jbc.m111.217935] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Insulin regulates glucose uptake through effects on the trafficking of the glucose transporter Glut4. To investigate the degree of overlap between Glut4 and the general endocytic pathways, the kinetics of trafficking of Glut4 and the receptors for transferrin (Tf) and α(2)-macroglobulin (α-2-M; LRP-1) were compared using quantitative flow cytometric assays. Insulin increased the exocytic rate constant (k(ex)) for both Glut4 and Tf. However, the k(ex) of Glut4 was 5-15 times slower than Tf in both basal and insulin-stimulated cells. The endocytic rate constant (k(en)) of Glut4 was also five times slower than Tf. Insulin did not affect the k(en) of either protein. In basal cells, the k(en) for α-2-M/LRP-1 was similar to Glut4 but 5-fold slower than Tf. Insulin increased k(en) for α-2-M/LRP-1 by 30%. In contrast, the k(ex) for LRP-1 was five times faster than Glut4 in basal cells, and insulin did not increase this rate constant. Thus, although there is overlap in the protein machineries/compartments utilized, the differences in trafficking kinetics indicate that Glut4, the Tf receptor, and LRP-1 are differentially processed both within the cell and at the plasma membrane. It has been reported that insulin decreases the k(en) of Glut4 in adipocytes. However, the effect of exocytosis on the "internalization" assays was not considered. Because it is counterintuitive, the effect of exocytosis on these assays is often overlooked in endocytosis studies. Using mathematical modeling and simulation, we show that the reported decrease in Glut4 k(en) can be entirely accounted for by the well established increase in Glut4 k(ex).
Collapse
Affiliation(s)
- Estifanos N Habtemichael
- Department of Biochemistry and Molecular Biology, University of Nevada School of Medicine, Reno, Nevada 89557, USA
| | | | | | | |
Collapse
|
30
|
Stenkula KG, Lizunov VA, Cushman SW, Zimmerberg J. Insulin controls the spatial distribution of GLUT4 on the cell surface through regulation of its postfusion dispersal. Cell Metab 2010; 12:250-9. [PMID: 20816091 PMCID: PMC3427691 DOI: 10.1016/j.cmet.2010.08.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Revised: 05/19/2010] [Accepted: 06/07/2010] [Indexed: 01/06/2023]
Abstract
While the glucose transporter-4 (GLUT4) is fundamental to insulin-regulated glucose metabolism, its dynamic spatial organization in the plasma membrane (PM) is unclear. Here, using multicolor TIRF microscopy in transfected adipose cells, we demonstrate that insulin regulates not only the exocytosis of GLUT4 storage vesicles but also PM distribution of GLUT4 itself. In the basal state, domains (clusters) of GLUT4 molecules in PM are created by an exocytosis that retains GLUT4 at the fusion site. Surprisingly, when insulin induces a burst of GLUT4 exocytosis, it does not merely accelerate this basal exocytosis but rather stimulates approximately 60-fold another mode of exocytosis that disperses GLUT4 into PM. In contradistinction, internalization of most GLUT4, regardless of insulin, occurs from pre-existing clusters via the subsequent recruitment of clathrin. The data fit a new kinetic model that features multifunctional clusters as intermediates of exocytosis and endocytosis.
Collapse
Affiliation(s)
- Karin G Stenkula
- Experimental Diabetes, Metabolism, and Nutrition Section, Diabetes Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
31
|
Amino acids influence the glucose uptake through GLUT4 in CHO-K1 cells under high glucose conditions. Mol Cell Biochem 2010; 344:43-53. [PMID: 20628794 DOI: 10.1007/s11010-010-0527-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Accepted: 06/22/2010] [Indexed: 01/11/2023]
Abstract
According to studies earlier, amino acids have proven to be antidiabetic, antiglycating, and anticataractogenic. The present study was to explore whether amino acids as mixtures could enhance glucose uptake in CHO-K1 cells specifically. The cells in F-12K1 serum-free medium were exposed to normal (7 mM) and high glucose (12, 17 and 27 mM) in the presence and absence of amino acids mixture (AAM) in varying concentration (2.5, 5 and 10 mM). The mixture 5 and 10 mM AAM increased the 2-deoxyglucose (2DG) uptake at all glucose concentration significantly. There was also a significant increase in the GLUT4 (glucose transporter) translocation as revealed by flow cytometer. Addition of a mixture of amino acids was found to improve cell viability, which got altered by high glucose in the CHO-K1 cells. Amino acids as mixture had a beneficial effect in improving the net utilization of glucose as an additive effect with insulin.
Collapse
|
32
|
Capilla E, Díaz M, Hou JC, Planas JV, Pessin JE. High basal cell surface levels of fish GLUT4 are related to reduced sensitivity of insulin-induced translocation toward GGA and AS160 inhibition in adipocytes. Am J Physiol Endocrinol Metab 2010; 298:E329-36. [PMID: 20075431 PMCID: PMC2822488 DOI: 10.1152/ajpendo.00547.2009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Glucose entry into cells is mediated by a family of facilitative transporter proteins (GLUTs). In mammals, GLUT4 is expressed in insulin-sensitive tissues and is responsible for the postprandial uptake of glucose. In fish, GLUT4 also mediates insulin-regulated glucose entry into cells but differs from mammalian GLUT4 in its affinity for glucose and in protein motifs known to be important for the traffic of GLUT4. In this study, we have characterized the intracellular and plasma membrane (PM) traffic of two orthologs of GLUT4 in fish, trout (btGLUT4) and salmon (okGLUT4), that do not share the amino terminal FQQI targeting motif of mammalian GLUT4. btGLUT4 (FQHL) and, to a lesser extent, okGLUT4 (FQQL) showed higher basal PM levels, faster traffic to the PM after biosynthesis, and earlier acquisition of insulin responsiveness than rat GLUT4. Furthermore, btGLUT4 showed a similar profile of internalization than rat GLUT4. Expression of the dominant-interfering AS160-4P mutant caused a significant decrease in the insulin-induced PM levels of okGLUT4 and rat GLUT4 and, to a lesser extent, of btGLUT4, suggesting that btGLUT4 has reduced retention into the IRC. Contrary to rat GLUT4 and okGLUT4, the presence of btGLUT4 at the PM under insulin-stimulated conditions was not affected by coexpression of a dominant-interfering GGA mutant. These data suggest that fish GLUT4 follow a different trafficking pathway to the PM compared with rat GLUT4 that seems to be relatively independent of GGA. These results indicate that the regulated trafficking characteristics of GLUT4 have been modified during evolution from fish to mammals.
Collapse
Affiliation(s)
- Encarnación Capilla
- Departament de Fisiologia, Facultat de Biologia, Universitat de Barcelona and Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | |
Collapse
|
33
|
Abstract
Genetic information flows from DNA to macromolecular structures-the dominant force in the molecular organization of life. However, recent work suggests that metabolite availability to the hexosamine and Golgi N-glycosylation pathways exerts control over the assembly of macromolecular complexes on the cell surface and, in this capacity, acts upstream of signaling and gene expression. The structure and number of N-glycans per protein molecule cooperate to regulate lectin binding and thereby the distribution of glycoproteins at the cell surface. Congenital disorders of glycosylation provide insight as extreme hypomorphisms, whereas milder deficiencies may encompass many common chronic conditions, including autoimmunity, metabolic syndrome, and aging.
Collapse
Affiliation(s)
- James W Dennis
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada.
| | | | | |
Collapse
|
34
|
Zhang ZF, Li Q, Liang J, Dai XQ, Ding Y, Wang JB, Li Y. Epigallocatechin-3-O-gallate (EGCG) protects the insulin sensitivity in rat L6 muscle cells exposed to dexamethasone condition. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2010; 17:14-18. [PMID: 19819682 DOI: 10.1016/j.phymed.2009.09.007] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 08/09/2009] [Accepted: 09/14/2009] [Indexed: 05/28/2023]
Abstract
The tea polyphenol epigallocatechin-3-O-gallate (EGCG) displays some antidiabetic effects; however the mechanisms are incompletely understood. In the present study, the investigation of the effects of EGCG on insulin resistance was performed in rat L6 cells treated with dexamethasone. We found that dexamethasone increased Ser307 phosphorylation of insulin receptor substrate-1 (IRS-1) and reduced phosphorylation of AMPK and Akt. Furthermore, glucose uptake and glucose transporter (GLUT4) translocation were inhibited by dexamethasone. However, the treatment of EGCG improved insulin-stimulated glucose uptake by increasing GLUT4 translocation to plasma membrane. Furthermore, we also demonstrated these EGCG effects essentially depended on the AMPK and Akt activation. Together, our data suggested that EGCG inhibited dexamethasone-induced insulin resistance through AMPK and PI3K/Akt pathway.
Collapse
Affiliation(s)
- Z F Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, PR China
| | | | | | | | | | | | | |
Collapse
|
35
|
GLUT4 molecules are recruited at random for insertion within the plasma membrane upon insulin stimulation. FEBS Lett 2009; 584:537-42. [DOI: 10.1016/j.febslet.2009.11.093] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Revised: 11/30/2009] [Accepted: 11/30/2009] [Indexed: 01/14/2023]
|
36
|
Fazakerley DJ, Holman GD, Marley A, James DE, Stöckli J, Coster ACF. Kinetic evidence for unique regulation of GLUT4 trafficking by insulin and AMP-activated protein kinase activators in L6 myotubes. J Biol Chem 2009; 285:1653-60. [PMID: 19915010 PMCID: PMC2804323 DOI: 10.1074/jbc.m109.051185] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
In L6 myotubes, redistribution of a hemagglutinin (HA) epitope-tagged GLUT4 (HA-GLUT4) to the cell surface occurs rapidly in response to insulin stimulation and AMP-activated protein kinase (AMPK) activation. We have examined whether these separate signaling pathways have a convergent mechanism that leads to GLUT4 mobilization and to changes in GLUT4 recycling. HA antibody uptake on GLUT4 in the basal steady state reached a final equilibrium level that was only 81% of the insulin-stimulated level. AMPK activators (5-aminoimidazole-4-carboxyamide ribonucleoside (AICAR) and A-769662) led to a similar level of antibody uptake to that found in insulin-stimulated cells. However, the combined responses to insulin stimulation and AMPK activation led to an antibody uptake level of ∼20% above the insulin level. Increases in antibody uptake due to insulin, but not AICAR or A-769662, treatment were reduced by both wortmannin and Akt inhibitor. The GLUT4 internalization rate constant in the basal steady state was very rapid (0.43 min−1) and was decreased during the steady-state responses to insulin (0.18 min−1), AICAR (0.16 min−1), and A-769662 (0.24 min−1). This study has revealed a nonconvergent mobilization of GLUT4 in response to activation of Akt and AMPK signaling. Furthermore, GLUT4 trafficking in L6 muscle cells is very reliant on regulated endocytosis for control of cell surface GLUT4 levels.
Collapse
Affiliation(s)
- Daniel J Fazakerley
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA27AY, United Kingdom
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
The association of receptors and solute transporters with components of the endocytic machinery regulates their surface levels, and thereby cellular sensitivity to cytokines, ligands and nutrients in the extracellular environment. Most transmembrane receptors and solute transporters are glycoproteins, and the Asn (N)-linked oligosaccharides (N-glycans) can bind animal lectins, forming multivalent lattices or microdomains that regulate glycoprotein mobility in the plane of membrane. The N-glycan number (sequence-encoded NXS/T) and context-dependent Golgi N-glycan branching cooperate to regulate glycoprotein affinities for the galectin family of lectins. Galectin-3 binding reduces EGF receptor trafficking into clathrin-coated pits and caveolae lipid rafts, decreases ligand-independent receptor activation and promotes alpha5beta1 integrin remodelling in focal adhesions. N-glycan branching in the medial Golgi increases glycan affinity for galectins, and the Golgi pathway is sensitive to uridine diphosphate-N-acetylglucosamine (UDP-GlcNAc) supply, in turn hexosamine pathway metabolites (fructose-6-P, glutamine and acetyl-CoA). Thus, lattice avidity and cellular responsiveness to extracellular cues are regulated in an adaptive manner by metabolism and Golgi modification to glycoproteins. Computational modelling of the hexosamine/Golgi/lattice has provided new insight on cell surface adaptation in cancer and autoimmune disease.
Collapse
Affiliation(s)
- James W Dennis
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, 600 University Avenue R988, Toronto, ON, Canada M5G 1X5.
| | | | | | | |
Collapse
|
38
|
Muretta JM, Mastick CC. How insulin regulates glucose transport in adipocytes. VITAMINS AND HORMONES 2009; 80:245-86. [PMID: 19251041 DOI: 10.1016/s0083-6729(08)00610-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Insulin stimulates glucose storage and metabolism by the tissues of the body, predominantly liver, muscle and fat. Storage in muscle and fat is controlled to a large extent by the rate of facilitative glucose transport across the plasma membrane of the muscle and fat cells. Insulin controls this transport. Exactly how remains debated. Work presented in this review focuses on the pathways responsible for the regulation of glucose transport by insulin. We present some historical work to show how the prevailing model for regulation of glucose transport by insulin was originally developed, then some more recent data challenging this model. We finish describing a unifying model for the control of glucose transport, and some very recent data illustrating potential molecular machinery underlying this regulation. This review is meant to give an overview of our current understanding of the regulation of glucose transport through the regulation of the trafficking of Glut4, highlighting important questions that remain to be answered. A more detailed treatment of specific aspects of this pathway can be found in several excellent recent reviews (Brozinick et al., 2007 Hou and Pessin, 2007; Huang and Czech, 2007;Larance et al., 2008 Sakamoto and Holman, 2008; Watson and Pessin, 2007; Zaid et al., 2008)One of the main objectives of this review is to discuss the results of the experiments measuring the kinetics of Glut4 movement between subcellular compartments in the context of our emerging model of the Glut4 trafficking pathway.
Collapse
Affiliation(s)
- Joseph M Muretta
- Department of Biochemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | |
Collapse
|
39
|
Compartmentalization and regulation of insulin signaling to GLUT4 by the cytoskeleton. VITAMINS AND HORMONES 2009; 80:193-215. [PMID: 19251039 DOI: 10.1016/s0083-6729(08)00608-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
One of the early events in the development of Type 2 diabetes appears to be an inhibition of insulin-mediated GLUT4 redistribution to the cell surface in tissues that express GLUT4. Understanding this process, and how it begins to breakdown in the development of insulin resistance is quite important as we face treatment and prevention of metabolic diseases. Over the past few years, and increasing number of laboratories have produced compelling data to demonstrate a role for both the actin and microtubule networks in the regulation of insulin-mediated GLUT4 redistribution to the cell surface. In this review, we explore this process from insulin-signal transduction to fusion of GLUT4 membrane vesicles, focusing on studies that have implicated a role for the cytoskeleton. We see from this body of work that both the actin network and the microtubule cytoskeleton play roles as targets of insulin action and effectors of insulin signaling leading to changes in GLUT4 redistribution to the cell surface and insulin-mediated glucose uptake.
Collapse
|
40
|
Rubin BR, Bogan JS. Intracellular retention and insulin-stimulated mobilization of GLUT4 glucose transporters. VITAMINS AND HORMONES 2009; 80:155-92. [PMID: 19251038 DOI: 10.1016/s0083-6729(08)00607-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
GLUT4 glucose transporters are expressed nearly exclusively in adipose and muscle cells, where they cycle to and from the plasma membrane. In cells not stimulated with insulin, GLUT4 is targeted to specialized GLUT4 storage vesicles (GSVs), which sequester it away from the cell surface. Insulin acts within minutes to mobilize these vesicles, translocating GLUT4 to the plasma membrane to enhance glucose uptake. The mechanisms controlling GSV sequestration and mobilization are poorly understood. An insulin-regulated aminopeptidase that cotraffics with GLUT4, IRAP, is required for basal GSV retention and insulin-stimulated mobilization. TUG and Ubc9 bind GLUT4, and likely retain GSVs within unstimulated cells. These proteins may be components of a retention receptor, which sequesters GLUT4 and IRAP away from recycling vesicles. Insulin may then act on this protein complex to liberate GLUT4 and IRAP, discharging GSVs into a recycling pathway for fusion at the cell surface. How GSVs are anchored intracellularly, and how insulin mobilizes these vesicles, are the important topics for ongoing research. Regulation of GLUT4 trafficking is tissue-specific, perhaps in part because the formation of GSVs requires cell type-specific expression of sortilin. Proteins controlling GSV retention and mobilization can then be more widely expressed. Indeed, GLUT4 likely participates in a general mechanism by which the cell surface delivery of various membrane proteins can be controlled by extracellular stimuli. Finally, it is not known if defects in the formation or intracellular retention of GSVs contribute to human insulin resistance, or play a role in the pathogenesis of type 2 diabetes.
Collapse
Affiliation(s)
- Bradley R Rubin
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520-8020, USA
| | | |
Collapse
|
41
|
Velebit J, Kovacic PB, Prebil M, Chowdhury HH, Grilc S, Kreft M, Jensen J, Isenović ER, Zorec R. Rosiglitazone modulates insulin-induced plasma membrane area changes in single 3T3-L1 adipocytes. J Membr Biol 2008; 223:141-9. [PMID: 18754069 DOI: 10.1007/s00232-008-9120-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Accepted: 07/08/2008] [Indexed: 11/30/2022]
Abstract
In this study we hypothesized that rosiglitazone, an antidiabetic high-affinity agonist for the peroxisome proliferator-activated receptor gamma, affects the plasma membrane (PM) turnover in single 3T3-L1 adipocytes. To study the PM turnover, the patch-clamp electrophysiological method was used to measure changes in membrane capacitance (Cm), a parameter linearly related to the PM area. Microscopy results show that the presence of rosiglitazone in the differentiating medium significantly increased the differentiation of 3T3-L1 adipocytes in cell culture, based on oil red O-stained area (11.4 +/- 1.2%) vs. controls (3.1 +/- 0.5%). Moreover, rosiglitazone treatment significantly reduced the size of single 3T3-L1 adipocytes; their average radius of 21.1 +/- 1.1 microm in controls was reduced to 17.5 +/- 0.5 microm in rosiglitazone-treated cells. Consistent with this, insulin application increased the rate of Cm increase to 2.34 +/- 0.10%/min, which was significantly different from controls (0.12 +/- 0.08%/min). However, pretreatment of cells with rosiglitazone prior to the treatment with insulin resulted in an attenuated rate of Cm increase. These data support the involvement of insulin in the modulation of membrane area and show that treatment by rosiglitazone reduced the insulin-mediated membrane area increase in 3T3-L1 adipocytes.
Collapse
Affiliation(s)
- Jelena Velebit
- Laboratory for Molecular Genetics and Radiobiology, Institute Vinca, P.O. Box 522, 11000 Belgrade, Serbia
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Randhawa VK, Ishikura S, Talior-Volodarsky I, Cheng AWP, Patel N, Hartwig JH, Klip A. GLUT4 vesicle recruitment and fusion are differentially regulated by Rac, AS160, and Rab8A in muscle cells. J Biol Chem 2008; 283:27208-19. [PMID: 18650435 DOI: 10.1074/jbc.m804282200] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Insulin increases glucose uptake into muscle by enhancing the surface recycling of GLUT4 transporters. In myoblasts, insulin signals bifurcate downstream of phosphatidylinositol 3-kinase into separate Akt and Rac/actin arms. Akt-mediated Rab-GAP AS160 phosphorylation and Rac/actin are required for net insulin gain of GLUT4, but the specific steps (vesicle recruitment, docking or fusion) regulated by Rac, actin dynamics, and AS160 target Rab8A are unknown. In L6 myoblasts expressing GLUT4myc, blocking vesicle fusion by tetanus toxin cleavage of VAMP2 impeded GLUT4myc membrane insertion without diminishing its build-up at the cell periphery. Conversely, actin disruption by dominant negative Rac or Latrunculin B abolished insulin-induced surface and submembrane GLUT4myc accumulation. Expression of non-phosphorylatable AS160 (AS160-4P) abrogated membrane insertion of GLUT4myc and partially reduced its cortical build-up, an effect magnified by selective Rab8A knockdown. We propose that insulin-induced actin dynamics participates in GLUT4myc vesicle retention beneath the membrane, whereas AS160 phosphorylation is essential for GLUT4myc vesicle-membrane docking/fusion and also contributes to GLUT4myc cortical availability through Rab8A.
Collapse
Affiliation(s)
- Varinder K Randhawa
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Recycling via the plasma membrane is a key feature that is shared by many membrane proteins. Using a combination of indirect immunofluorescence labeling and fluorescence detection using a fluorescence multiwell plate reader, we exploited the possibilities of quantitatively measuring the trafficking kinetics of transmembrane proteins. Parameters that can be studied include dynamic appearance/presence at the cell surface, recycling via the cell surface, and internalization. For the insulin-responsive glucose transporter GLUT4 (glucose transporter number 4), details are presented on how to quantitatively measure insulin-induced GLUT4 translocation toward the plasma membrane (transition state) and to analyze cell surface recycling of GLUT4 in basal and insulin-stimulated cells (steady state).
Collapse
|
44
|
Muretta JM, Romenskaia I, Mastick CC. Insulin releases Glut4 from static storage compartments into cycling endosomes and increases the rate constant for Glut4 exocytosis. J Biol Chem 2007; 283:311-323. [PMID: 17967900 DOI: 10.1074/jbc.m705756200] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
In adipocytes, insulin triggers the redistribution of Glut4 from intracellular compartments to the plasma membrane. Two models have been proposed to explain the effect of insulin on Glut4 localization. In the first, termed dynamic exchange, Glut4 continually cycles between the plasma membrane and intracellular compartments in basal cells, and the major effect of insulin is through changes in the exocytic and endocytic rate constants, k(ex) and k(en). In the second model, termed static retention, Glut4 is packaged in specialized storage vesicles (GSVs) in basal cells and does not traffic through the plasma membrane or endosomes. Insulin triggers GSV exocytosis, increasing the amount of Glut4 in the actively cycling pool. Using a flow cytometry-based assay, we found that Glut4 is regulated by both static and dynamic retention mechanisms. In basal cells, 75-80% of the Glut4 is packaged in noncycling GSVs. Insulin increased the amount of Glut4 in the actively cycling pool 4-5-fold. Insulin also increased k(ex) in the cycling pool 3-fold. After insulin withdrawal, Glut4 is rapidly cleared from the plasma membrane (t((1/2)) of 20 min) by rapid adjustments in k(ex) and k(en) and recycled into static compartments. Complete recovery of the static pool required more than 3 h, however. We conclude that in fully differentiated confluent adipocytes, both the dynamic and static retention mechanisms are important for the regulation of plasma membrane Glut4 content. However, cell culture conditions affect Glut4 trafficking. For example, replating after differentiation inhibited the static retention of Glut4, which may explain differences in previous reports.
Collapse
Affiliation(s)
- Joseph M Muretta
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Nevada 89557
| | - Irina Romenskaia
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Nevada 89557
| | - Cynthia Corley Mastick
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Nevada 89557.
| |
Collapse
|
45
|
Liu LB, Omata W, Kojima I, Shibata H. The SUMO conjugating enzyme Ubc9 is a regulator of GLUT4 turnover and targeting to the insulin-responsive storage compartment in 3T3-L1 adipocytes. Diabetes 2007; 56:1977-85. [PMID: 17536066 DOI: 10.2337/db06-1100] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The small ubiquitin-related modifier (SUMO) conjugating enzyme Ubc9 has been shown to upregulate GLUT4 in L6 myoblast cells, although the mechanism of action has remained undefined. Here we investigated the physiological significance of Ubc9 in GLUT4 turnover and subcellular targeting by adenovirus vector-mediated overexpression and by small interfering RNA (siRNA)-mediated gene silencing of Ubc9 in 3T3-L1 adipocytes. Overexpression of Ubc9 resulted in an inhibition of GLUT4 degradation and promoted its targeting to the unique insulin-responsive GLUT4 storage compartment (GSC), leading to an increase in GLUT4 amount and insulin-responsive glucose transport in 3T3-L1 adipocytes. Overexpression of Ubc9 also antagonized GLUT4 downregulation and its selective loss in GSC induced by long-term insulin stimulation. By contrast, siRNA-mediated depletion of Ubc9 accelerated GLUT4 degradation and decreased the amount of the transporter, concurrent with its selective loss in GSC, which resulted in attenuated insulin-responsive glucose transport. Intriguingly, overexpression of the catalytically inactive mutant Ubc9-C93A produced effects indistinguishable from those with wild-type Ubc9, suggesting that Ubc9 regulates GLUT4 turnover and targeting to GSC by a mechanism independent of its catalytic activity. Thus, Ubc9 is a pivotal regulator of the insulin sensitivity of glucose transport in adipocytes.
Collapse
Affiliation(s)
- Li-Bin Liu
- Department of Cell Biology, Institute for Molecular and Cellular Regulation, Gunma University, Showa-machi, Maebashi, Japan
| | | | | | | |
Collapse
|
46
|
Hou JC, Pessin JE. Ins (endocytosis) and outs (exocytosis) of GLUT4 trafficking. Curr Opin Cell Biol 2007; 19:466-73. [PMID: 17644329 PMCID: PMC2041936 DOI: 10.1016/j.ceb.2007.04.018] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2007] [Accepted: 04/17/2007] [Indexed: 12/21/2022]
Abstract
Glucose transporter 4 (GLUT4) is the major insulin-regulated glucose transporter expressed mainly in muscle and adipose tissue. GLUT4 is stored in a poorly characterized intracellular vesicular compartment and translocates to the cell surface in response to insulin stimulation resulting in an increased glucose uptake. This process is essential for the maintenance of normal glucose homeostasis and involves a complex interplay of trafficking events and intracellular signaling cascades. Recent studies have identified sortilin as an essential element for the formation of GLUT4 storage vesicles during adipogenesis and Golgi-localized gamma-ear-containing Arf-binding protein (GGA) as a key coat adaptor for the entry of newly synthesized GLUT4 into the specialized compartment. Insulin-stimulated GLUT4 translocation from this compartment to the plasma membrane appears to require the Akt/protein kinase B substrate termed AS160 (Akt substrate of 160kDa). In addition, the VPS9 domain-containing protein Gapex-5 in complex with CIP4 appears to function as a Rab31 guanylnucleotide exchange factor that is necessary for insulin-stimulated GLUT4 translocation. Here, we attempt to summarize recent advances in GLUT4 vesicle biogenesis, intracellular trafficking and membrane fusion.
Collapse
Affiliation(s)
- June Chunqiu Hou
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA.
| | | |
Collapse
|
47
|
Fernando RN, Luff SE, Albiston AL, Chai SY. Sub-cellular localization of insulin-regulated membrane aminopeptidase, IRAP to vesicles in neurons. J Neurochem 2007; 102:967-76. [PMID: 17504262 DOI: 10.1111/j.1471-4159.2007.04659.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Angiotensin IV and LVV-hemorphin 7 promote robust enhancing effects on learning and memory. These peptides are also competitive inhibitors of the insulin-regulated membrane aminopeptidase, suggesting that the biological actions of these peptides may result from inhibition of IRAP activity. However, the normal function of IRAP in the brain is yet to be determined. The present study investigated the sub-cellular distribution of IRAP in four neuronal cell lines and in the mouse brain. Using sub-cellular fractionation, IRAP was found to be enriched in low density microsomes, while lower levels of IRAP were also present in high density microsomes, plasma membrane and mitochondrial fractions. Dual-label immunohistochemistry confirmed the presence of IRAP in vesicles co-localized with the vesicular maker VAMP2, in the trans Golgi network co-localized with TGN 38 and in endosomes co-localized with EEA1. Finally using electron microscopy, IRAP specific immunoreactivity was predominantly associated with large 100-200 nm vesicles in hippocampal neurons. The location, appearance and size of these vesicles are consistent with neurosecretory vesicles. IRAP precipitate was also detected in intracellular structures including the rough endoplasmic reticulum, Golgi stack and mitochondrial membranes. The sub-cellular localization of IRAP in neurons demonstrated in the present study bears striking parallels with distribution of IRAP in insulin responsive cells, where the enzyme plays a role in insulin-regulated glucose uptake. Therefore, we propose that the function of IRAP in neurons may be similar to that in insulin responsive cells.
Collapse
Affiliation(s)
- Ruani N Fernando
- Howard Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| | | | | | | |
Collapse
|
48
|
Abstract
Few physiological parameters are more tightly and acutely regulated in humans than blood glucose concentration. The major cellular mechanism that diminishes blood glucose when carbohydrates are ingested is insulin-stimulated glucose transport into skeletal muscle. Skeletal muscle both stores glucose as glycogen and oxidizes it to produce energy following the transport step. The principal glucose transporter protein that mediates this uptake is GLUT4, which plays a key role in regulating whole body glucose homeostasis. This review focuses on recent advances on the biology of GLUT4.
Collapse
Affiliation(s)
- Shaohui Huang
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | |
Collapse
|
49
|
Yu C, Cresswell J, Löffler MG, Bogan JS. The glucose transporter 4-regulating protein TUG is essential for highly insulin-responsive glucose uptake in 3T3-L1 adipocytes. J Biol Chem 2007; 282:7710-22. [PMID: 17202135 PMCID: PMC2243252 DOI: 10.1074/jbc.m610824200] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Insulin stimulates glucose uptake in fat and muscle by redistributing GLUT4 glucose transporters from intracellular membranes to the cell surface. We previously proposed that, in 3T3-L1 adipocytes, TUG retains GLUT4 within unstimulated cells and insulin mobilizes this retained GLUT4 by stimulating its dissociation from TUG. Yet the relative importance of this action in the overall control of glucose uptake remains uncertain. Here we report that transient, small interfering RNA-mediated depletion of TUG causes GLUT4 translocation and enhances glucose uptake in unstimulated 3T3-L1 adipocytes, similar to insulin. Stable TUG depletion or expression of a dominant negative fragment likewise stimulates GLUT4 redistribution and glucose uptake, and insulin causes a 2-fold further increase. Microscopy shows that TUG governs the accumulation of GLUT4 in perinuclear membranes distinct from endosomes and indicates that it is this pool of GLUT4 that is mobilized by TUG disruption. Interestingly, in addition to translocating GLUT4 and enhancing glucose uptake, TUG disruption appears to accelerate the degradation of GLUT4 in lysosomes. Finally, we find that TUG binds directly and specifically to a large intracellular loop in GLUT4. Together, these findings demonstrate that TUG is required to retain GLUT4 intracellularly in 3T3-L1 adipocytes in the absence of insulin and further implicate the insulin-stimulated dissociation of TUG and GLUT4 as an important action by which insulin stimulates glucose uptake.
Collapse
Affiliation(s)
- Chenfei Yu
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - James Cresswell
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Michael G. Löffler
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Jonathan S. Bogan
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520
- ¶Address correspondence to: Jonathan S. Bogan, Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar St., P.O. Box 208020, New Haven, CT 06520-8020, Tel. (203) 785-6319; Fax. (203) 785-6462; E-Mail:
| |
Collapse
|
50
|
Abstract
Previous studies have suggested that activation of the Rho family member GTPase TC10 is necessary but not sufficient for the stimulation of glucose transport by insulin. We show here that endogenous TC10alpha is rapidly activated in response to insulin in 3T3L1 adipocytes in a phosphatidylinositol 3-kinase-independent manner, whereas platelet-derived growth factor was without effect. Knockdown of TC10alpha but not TC10beta by RNA interference inhibited insulin-stimulated glucose uptake as well as the translocation of the insulin-sensitive glucose transporter GLUT4 from intracellular sites to the plasma membrane. In contrast, loss of TC10alpha had no effect on the stimulation of Akt by insulin. Additionally, knockdown of TC10alpha inhibited insulin-stimulated translocation of its effector CIP4. These data indicate that TC10alpha is specifically required for insulin-stimulated glucose uptake in adipocytes.
Collapse
Affiliation(s)
- Louise Chang
- Life Sciences Institute, University of Michigan Medical Center, Ann Arbor, Michigan 48109-2216, USA
| | | | | |
Collapse
|