1
|
Wang Y, Spolitu S, Zadroga JA, Sarecha AK, Ozcan L. Hepatocyte Rap1a contributes to obesity- and statin-associated hyperglycemia. Cell Rep 2022; 40:111259. [PMID: 36001955 PMCID: PMC9446800 DOI: 10.1016/j.celrep.2022.111259] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 05/26/2022] [Accepted: 08/03/2022] [Indexed: 12/28/2022] Open
Abstract
Excessive hepatic glucose production contributes to the development of hyperglycemia and is a key feature of type 2 diabetes. Here, we report that activation of hepatocyte Rap1a suppresses gluconeogenic gene expression and glucose production, whereas Rap1a silencing stimulates them. Rap1a activation is suppressed in obese mouse liver, and restoring its activity improves glucose intolerance. As Rap1a′s membrane localization and activation depends on its geranylgeranylation, which is inhibited by statins, we show that statin-treated hepatocytes and the human liver have lower active-Rap1a levels. Similar to Rap1a inhibition, statins stimulate hepatic gluconeogenesis and increase fasting blood glucose in obese mice. Geranylgeraniol treatment, which acts as the precursor for geranylgeranyl isoprenoids, restores Rap1a activity and improves statin-mediated glucose intolerance. Mechanistically, Rap1a activation induces actin polymerization, which suppresses gluconeogenesis by Akt-mediated FoxO1 inhibition. Thus, Rap1a regulates hepatic glucose homeostasis, and blocking its activity, via lowering geranylgeranyl isoprenoids, contributes to statin-induced glucose intolerance. Wang et al. show that activation of hepatic Rap1a suppresses gluconeogenic gene expression and improves glucose intolerance via Akt-mediated FoxO1 inhibition. Statins lower intracellular isoprenoid levels and inhibit Rap1a activation, which contributes to their hyperglycemic effect. These findings identify a role of hepatic Rap1a in obesity- and statin-associated glucose homeostasis.
Collapse
Affiliation(s)
- Yating Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Stefano Spolitu
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - John A Zadroga
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Amesh K Sarecha
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Lale Ozcan
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
2
|
Dogra S, Neelakantan D, Patel MM, Griesel B, Olson A, Woo S. Adipokine Apelin/APJ Pathway Promotes Peritoneal Dissemination of Ovarian Cancer Cells by Regulating Lipid Metabolism. Mol Cancer Res 2021; 19:1534-1545. [PMID: 34172534 PMCID: PMC11486291 DOI: 10.1158/1541-7786.mcr-20-0991] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 04/05/2021] [Accepted: 05/11/2021] [Indexed: 11/16/2022]
Abstract
Adipose tissue, which can provide adipokines and nutrients to tumors, plays a key role in promoting ovarian cancer metastatic lesions in peritoneal cavity. The adipokine apelin promotes ovarian cancer metastasis and progression through its receptor APJ, which regulates cell proliferation, energy metabolism, and angiogenesis. The objective of this study was to investigate the functional role and mechanisms of the apelin-APJ pathway in ovarian cancer metastasis, especially in context of tumor cell-adipocyte interactions. When co-cultured in the conditioned media (AdipoCM) derived from 3T3-L1 adipocytes, which express and secrete high apelin, human ovarian cancer cells with high APJ expression showed significant increases in migration and invasion in vitro. We also found that cells expressing high levels of APJ had increased cell adhesion to omentum ex vivo, and preferentially "home-in" on the omentum in vivo. These apelin-induced pro-metastatic effects were reversed by APJ antagonist F13A in a dose-dependent manner. Apelin-APJ activation increased lipid droplet accumulation in ovarian cancer cells, which was further intensified in the presence of AdipoCM and reversed by F13A or APJ knockdown. Mechanistically, this increased lipid uptake was mediated by CD36 upregulation via APJ-STAT3 activation, and the lipids were utilized in promoting fatty acid oxidation via activation of AMPK-CPT1a axis. Together, our studies demonstrate that adipocyte-derived apelin activates APJ-expressing tumor cells in a paracrine manner, promoting lipid uptake and utilization and providing energy for ovarian cancer cell survival at the metastatic sites. Hence, the apelin-APJ pathway presents a novel therapeutic target to curb ovarian cancer metastasis. IMPLICATIONS: Targeting the APJ pathway in high-grade serous ovarian carcinoma is a novel strategy to inhibit peritoneal metastasis.
Collapse
Affiliation(s)
- Samrita Dogra
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Deepika Neelakantan
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Maulin M Patel
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Cardiovascular Biology Department, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Beth Griesel
- Department of Biochemistry and Molecular Biology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Ann Olson
- Department of Biochemistry and Molecular Biology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Sukyung Woo
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, New York
| |
Collapse
|
3
|
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
|
4
|
Jackson RM, Griesel BA, Short KR, Sparling D, Freeman WM, Olson AL. Weight Loss Results in Increased Expression of Anti-Inflammatory Protein CRISPLD2 in Mouse Adipose Tissue. Obesity (Silver Spring) 2019; 27:2025-2036. [PMID: 31746554 PMCID: PMC6873817 DOI: 10.1002/oby.22652] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 08/16/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Obesity is a major risk factor for cardiovascular disease, metabolic syndrome, and type 2 diabetes mellitus, whereas weight loss is associated with improved health outcomes. It is therefore important to learn how adipose contraction during weight loss contributes to improved health. It was hypothesized that adipose tissue undergoing weight loss would have a unique transcriptomic profile, expressing specific genes that might improve health. METHODS This study conducted an RNA-sequencing analysis of the epididymal adipose tissue of mice fed either a high-fat diet (HFD) or a regular rodent chow diet (RD) ad libitum for 10 weeks versus a cohort of mice fed HFD for the first 5 weeks before being swapped to an RD for the remainder of the study (swapped diet [SWAP]). RESULTS The swapped diet resulted in weight loss, with a parallel improvement in insulin sensitivity. RNA sequencing revealed several transcriptomic signatures distinct to adipose tissue in SWAP mice, distinguished from both RD and HFD adipose tissue. The analysis found a unique upregulated mRNA that encodes a secreted lipopolysaccharide-binding glycoprotein (CRISPLD2) in adipose tissue. Whereas cellular CRISPLD2 protein levels were unchanged, plasma CRIPSLD2 levels increased in SWAP mice following weight loss and could correlate with insulin sensitivity. CONCLUSIONS Taken together, these data demonstrate that CRISPLD2 is a circulating adipokine that may regulate adipocyte remodeling during weight loss.
Collapse
Affiliation(s)
- Robert M. Jackson
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Beth A. Griesel
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Kevin R. Short
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - David Sparling
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Willard M. Freeman
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Ann Louise Olson
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
5
|
Masters TA, Tumbarello DA, Chibalina MV, Buss F. MYO6 Regulates Spatial Organization of Signaling Endosomes Driving AKT Activation and Actin Dynamics. Cell Rep 2018; 19:2088-2101. [PMID: 28591580 PMCID: PMC5469940 DOI: 10.1016/j.celrep.2017.05.048] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/05/2017] [Accepted: 05/12/2017] [Indexed: 02/06/2023] Open
Abstract
APPL1- and RAB5-positive signaling endosomes play a crucial role in the activation of AKT in response to extracellular stimuli. Myosin VI (MYO6) and two of its cargo adaptor proteins, GIPC and TOM1/TOM1L2, localize to these peripheral endosomes and mediate endosome association with cortical actin filaments. Loss of MYO6 leads to the displacement of these endosomes from the cell cortex and accumulation in the perinuclear space. Depletion of this myosin not only affects endosome positioning, but also induces actin and lipid remodeling consistent with endosome maturation, including accumulation of F-actin and the endosomal lipid PI(3)P. These processes acutely perturb endosome function, as both AKT phosphorylation and RAC-dependent membrane ruffling were markedly reduced by depletion of either APPL1 or MYO6. These results place MYO6 and its binding partners at a central nexus in cellular signaling linking actin dynamics at the cell surface and endosomal signaling in the cell cortex.
Collapse
Affiliation(s)
- Thomas A Masters
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Hills Road, Cambridge CB2 0XY, UK
| | - David A Tumbarello
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Hills Road, Cambridge CB2 0XY, UK
| | - Margarita V Chibalina
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Hills Road, Cambridge CB2 0XY, UK
| | - Folma Buss
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Hills Road, Cambridge CB2 0XY, UK.
| |
Collapse
|
6
|
Translocation and Redistribution of GLUT4 Using a Dual-Labeled Reporter Assay. Methods Mol Biol 2017. [PMID: 29218525 DOI: 10.1007/978-1-4939-7507-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
It is crucial to determine the regulation of GLUT4 translocation and redistribution to the plasma membrane. The HA-GLUT4-GFP dual-reporter construct has become an important tool in the assessment of GLUT4 recycling in cultured adipocytes and myocytes. Through the use of light microscopy, this reporter construct allows for visualization of GLUT4 specifically at the cell surface or GLUT4 that has recycled from the cell surface while simultaneously marking the total GLUT4 pool. Here, we discuss and outline the general application of this reporter construct and its use in evaluating GLUT4 translocation within cultured adipocytes.
Collapse
|
7
|
Jackson RM, Griesel BA, Gurley JM, Szweda LI, Olson AL. Glucose availability controls adipogenesis in mouse 3T3-L1 adipocytes via up-regulation of nicotinamide metabolism. J Biol Chem 2017; 292:18556-18564. [PMID: 28916720 DOI: 10.1074/jbc.m117.791970] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 09/13/2017] [Indexed: 12/21/2022] Open
Abstract
Expansion of adipose tissue in response to a positive energy balance underlies obesity and occurs through both hypertrophy of existing cells and increased differentiation of adipocyte precursors (hyperplasia). To better understand the nutrient signals that promote adipocyte differentiation, we investigated the role of glucose availability in regulating adipocyte differentiation and maturation. 3T3-L1 preadipocytes were grown and differentiated in medium containing a standard differentiation hormone mixture and either 4 or 25 mm glucose. Adipocyte maturation at day 9 post-differentiation was determined by key adipocyte markers, including glucose transporter 4 (GLUT4) and adiponectin expression and Oil Red O staining of neutral lipids. We found that adipocyte differentiation and maturation required a pulse of 25 mm glucose only during the first 3 days of differentiation. Importantly, fatty acids were unable to substitute for the 25 mm glucose pulse during this period. The 25 mm glucose pulse increased adiponectin and GLUT4 expression and accumulation of neutral lipids via distinct mechanisms. Adiponectin expression and other early markers of differentiation required an increase in the intracellular pool of total NAD/P. In contrast, GLUT4 protein expression was only partially restored by increased NAD/P levels. Furthermore, GLUT4 mRNA expression was mediated by glucose-dependent activation of GLUT4 gene transcription through the cis-acting GLUT4-liver X receptor element (LXRE) promoter element. In summary, this study supports the conclusion that high glucose promotes adipocyte differentiation via distinct metabolic pathways and independently of fatty acids. This may partly explain the mechanism underlying adipocyte hyperplasia that occurs much later than adipocyte hypertrophy in the development of obesity.
Collapse
Affiliation(s)
- Robert M Jackson
- From the Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104 and
| | - Beth A Griesel
- From the Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104 and
| | - Jami M Gurley
- From the Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104 and
| | - Luke I Szweda
- From the Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104 and.,the Program in Aging and Metabolism, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| | - Ann Louise Olson
- the Program in Aging and Metabolism, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| |
Collapse
|
8
|
Kido K, Yokokawa T, Ato S, Sato K, Fujita S. Effect of resistance exercise under conditions of reduced blood insulin on AMPKα Ser485/491 inhibitory phosphorylation and AMPK pathway activation. Am J Physiol Regul Integr Comp Physiol 2017; 313:R110-R119. [PMID: 28515080 DOI: 10.1152/ajpregu.00063.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/02/2017] [Accepted: 05/15/2017] [Indexed: 11/22/2022]
Abstract
Insulin stimulates skeletal muscle glucose uptake via activation of the protein kinase B/Akt (Akt) pathway. Recent studies suggest that insulin downregulates AMP-activated protein kinase (AMPK) activity via Ser485/491 phosphorylation of the AMPK α-subunit. Thus lower blood insulin concentrations may induce AMPK signal activation. Acute exercise is one method to stimulate AMPK activation; however, no study has examined the relationship between blood insulin levels and acute resistance exercise-induced AMPK pathway activation. Based on previous findings, we hypothesized that the acute resistance exercise-induced AMPK pathway activation would be augmented by disruptions in insulin secretion through a decrease in AMPKα Ser485/491 inhibitory phosphorylation. To test the hypothesis, 10-wk-old male Sprague-Dawley rats were administered the toxin streptozotocin (STZ; 55 mg/kg) to destroy the insulin secreting β-cells. Three days postinjection, the right gastrocnemius muscle from STZ and control rats was subjected to resistance exercise by percutaneous electrical stimulation. Animals were killed 0, 1, or 3 h later; activation of the Akt/AMPK and downstream pathways in the muscle tissue was analyzed by Western blotting and real-time PCR. Notably, STZ rats showed a significant decrease in basal Akt and AMPKα Ser485/491 phosphorylation, but substantial exercise-induced increases in both AMPKα Thr172 and acetyl-CoA carboxylase (ACC) Ser79 phosphorylation were observed. Although no significant impact on resistance exercise-induced Akt pathway activation or glucose uptake was found, resistance exercise-induced peroxisome proliferator-activated receptor (PPAR)-γ coactivator-1 α (PGC-1α) gene expression was augmented by STZ treatment. Collectively, these data suggest that circulating insulin levels may regulate acute resistance exercise-induced AMPK pathway activation and AMPK-dependent gene expression relating to basal AMPKα Ser485/491 phosphorylation.
Collapse
Affiliation(s)
- Kohei Kido
- Faculty of Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Takumi Yokokawa
- Laboratory of Sports and Exercise Medicine, Graduate School of Human and Environmental Studies, Kyoto University, Kyoto, Japan; and
| | - Satoru Ato
- Faculty of Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Koji Sato
- Graduate School of Human Development and Environment, Kobe University, Kobe, Hyogo, Japan
| | - Satoshi Fujita
- Faculty of Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga, Japan;
| |
Collapse
|
9
|
Increase in tumor suppressor Arf compensates gene dysregulation in in vitro aged adipocytes. Biogerontology 2016; 18:55-68. [DOI: 10.1007/s10522-016-9661-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/19/2016] [Indexed: 10/20/2022]
|
10
|
Yang S, Zhou L, Reilly PT, Shen SM, He P, Zhu XN, Li CX, Wang LS, Mak TW, Chen GQ, Yu Y. ANP32B deficiency impairs proliferation and suppresses tumor progression by regulating AKT phosphorylation. Cell Death Dis 2016; 7:e2082. [PMID: 26844697 PMCID: PMC4849165 DOI: 10.1038/cddis.2016.8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/31/2016] [Accepted: 01/04/2016] [Indexed: 01/10/2023]
Abstract
The acidic leucine-rich nuclear phosphoprotein 32B (ANP32B) is reported to impact normal development, with Anp32b-knockout mice exhibiting smaller size and premature aging. However, its cellular and molecular mechanisms, especially its potential roles in tumorigenesis, remain largely unclear. Here, we utilize 'knockout' models, RNAi silencing and clinical cohorts to more closely investigate the role of this enigmatic factor in cell proliferation and cancer phenotypes. We report that, compared with Anp32b wild-type (Anp32b+/+) littermates, a broad panel of tissues in Anp32b-deficient (Anp32b−/−) mice are demonstrated hypoplasia. Anp32b−/− mouse embryo fibroblast cell has a slower proliferation, even after oncogenic immortalization. ANP32B knockdown also significantly inhibits in vitro and in vivo growth of cancer cells by inducing G1 arrest. In line with this, ANP32B protein has higher expression in malignant tissues than adjacent normal tissues from a cohort of breast cancer patients, and its expression level positively correlates with their histopathological grades. Moreover, ANP32B deficiency downregulates AKT phosphorylation, which involves its regulating effect on cell growth. Collectively, our findings suggest that ANP32B is an oncogene and a potential therapeutic target for breast cancer treatment.
Collapse
Affiliation(s)
- S Yang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - L Zhou
- Department of Surgery, Branch of Shanghai First People's Hospital, SJTU-SM, Shanghai, China
| | - P T Reilly
- Laboratory of Inflammation Biology, National Cancer Centre Singapore, Singapore, Singapore
| | - S-M Shen
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - P He
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - X-N Zhu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - C-X Li
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - L-S Wang
- State Key Laboratory of Genetic Engineering, Minhang Hospital, Fudan University, Shanghai, China
| | - T W Mak
- Campbell Family Cancer Research Institute, University Health Network, Toronto, ON, Canada
| | - G-Q Chen
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Y Yu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine (SJTU-SM), Shanghai, China
| |
Collapse
|
11
|
An optogenetic system for interrogating the temporal dynamics of Akt. Sci Rep 2015; 5:14589. [PMID: 26423353 PMCID: PMC4589684 DOI: 10.1038/srep14589] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 09/02/2015] [Indexed: 11/30/2022] Open
Abstract
The dynamic activity of the serine/threonine kinase Akt is crucial for the regulation of diverse cellular functions, but the precise spatiotemporal control of its activity remains a critical issue. Herein, we present a photo-activatable Akt (PA-Akt) system based on a light-inducible protein interaction module of Arabidopsis thaliana cryptochrome2 (CRY2) and CIB1. Akt fused to CRY2phr, which is a minimal light sensitive domain of CRY2 (CRY2-Akt), is reversibly activated by light illumination in several minutes within a physiological dynamic range and specifically regulates downstream molecules and inducible biological functions. We have generated a computational model of CRY2-Akt activation that allows us to use PA-Akt to control the activity quantitatively. The system provides evidence that the temporal patterns of Akt activity are crucial for generating one of the downstream functions of the Akt-FoxO pathway; the expression of a key gene involved in muscle atrophy (Atrogin-1). The use of an optical module with computational modeling represents a general framework for interrogating the temporal dynamics of biomolecules by predictive manipulation of optogenetic modules.
Collapse
|
12
|
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
|
13
|
Sylow L, Kleinert M, Pehmøller C, Prats C, Chiu TT, Klip A, Richter EA, Jensen TE. Akt and Rac1 signaling are jointly required for insulin-stimulated glucose uptake in skeletal muscle and downregulated in insulin resistance. Cell Signal 2014; 26:323-31. [DOI: 10.1016/j.cellsig.2013.11.007] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 11/01/2013] [Indexed: 11/16/2022]
|
14
|
Lee A, Hakuno F, Northcott P, Pessin JE, Adcock MR. Nexilin, a cardiomyopathy-associated F-actin binding protein, binds and regulates IRS1 signaling in skeletal muscle cells. PLoS One 2013; 8:e55634. [PMID: 23383252 PMCID: PMC3559603 DOI: 10.1371/journal.pone.0055634] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 01/03/2013] [Indexed: 01/11/2023] Open
Abstract
Insulin stimulates glucose uptake through a highly organized and complex process that involves movement of the glucose transporter 4 (GLUT4) from intracellular storage sites to the plasma membrane. Previous studies in L6 skeletal muscle cells have shown that insulin-induced activation and assembly of insulin receptor substrate 1 (IRS1) and p85α the regulatory subunit of the Type 1A phosphatidylinositol-3-kinase (PI3K), within remodeled actin-rich membrane structures is critical for downstream signalling mediating the translocation of GLUT4. The mechanism for localization within actin cytoskeletal scaffolds is not known, as direct interaction of IRS1 or p85α with F-actin has not been demonstrated. Here we show that nexilin, a F-actin binding protein implicated in the pathogenesis of familial dilated cardiomyopathies, preferentially binds to IRS1 over IRS2 to influence glucose transport in skeletal muscle cells. Nexilin stably associates with IRS1 under basal conditions in L6 myotubes and this complex is disassembled by insulin. Exposure of L6 myotubes to Latrunculin B disrupts the spatial patterning of nexilin and its transient association with IRS1. Functional silencing of nexilin has no effect on insulin-stimulated IRS1 tyrosine phosphorylation, however it enhances recruitment of p85α to IRS1 resulting in increased PI-3, 4, 5-P3 formation, coincident with enhanced AKT activation and glucose uptake. By contrast, overexpression of nexilin inhibits transmission of IRS1 signals to AKT. Based on these findings we propose that nexilin may tether IRS1 to actin-rich structures under basal conditions, confining IRS1 signaling to specific subcellular locations in the cell. Insulin-elicited release of this constraint may enhance the efficiency of IRS1/PI3K interaction and PI-3, 4, 5-P3 production at localized sites. Moreover, the selective binding of nexilin to IRS1 and not IRS2 may contribute to the differential specificity of IRS isoforms in the modulation of GLUT4 trafficking in skeletal muscle cells.
Collapse
Affiliation(s)
- Andrew Lee
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Fumihiko Hakuno
- Department of Animal Sciences and Applied Biological Chemistry, The University of Tokyo, Tokyo, Japan
| | - Paul Northcott
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Jeffrey E. Pessin
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Maria Rozakis Adcock
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
15
|
Langlais P, Dillon JL, Mengos A, Baluch DP, Ardebili R, Miranda DN, Xie X, Heckmann BL, Liu J, Mandarino LJ. Identification of a role for CLASP2 in insulin action. J Biol Chem 2012; 287:39245-53. [PMID: 22992739 DOI: 10.1074/jbc.m112.394148] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Insulin stimulates the mobilization of glucose transporter 4 (GLUT4) storage vesicles to the plasma membrane, resulting in an influx of glucose into target tissues such as muscle and fat. We present evidence that CLIP-associating protein 2 (CLASP2), a protein previously unassociated with insulin action, is responsive to insulin stimulation. Using mass spectrometry-based protein identification combined with phosphoantibody immunoprecipitation in L6 myotubes, we detected a 4.8-fold increase of CLASP2 in the anti-phosphoserine immunoprecipitates upon insulin stimulation. Western blotting of CLASP2 immunoprecipitates with the phosphoantibody confirmed the finding that CLASP2 undergoes insulin-stimulated phosphorylation, and a number of novel phosphorylation sites were identified. Confocal imaging of L6 myotubes revealed that CLASP2 colocalizes with GLUT4 at the plasma membrane within areas of insulin-mediated cortical actin remodeling. CLASP2 is responsible for directing the distal end of microtubules to the cell cortex, and it has been shown that GLUT4 travels along microtubule tracks. In support of the concept that CLASP2 plays a role in the trafficking of GLUT4 at the cell periphery, CLASP2 knockdown by siRNA in L6 myotubes interfered with insulin-stimulated GLUT4 localization to the plasma membrane. Furthermore, siRNA mediated knockdown of CLASP2 in 3T3-L1 adipocytes inhibited insulin-stimulated glucose transport. We therefore propose a new model for CLASP2 in insulin action, where CLASP2 directs the delivery of GLUT4 to cell cortex landing zones important for insulin action.
Collapse
Affiliation(s)
- Paul Langlais
- Center for Metabolic and Vascular Biology, Arizona State University, Tempe, Arizona 85287, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Balamatsias D, Kong AM, Waters JE, Sriratana A, Gurung R, Bailey CG, Rasko JEJ, Tiganis T, Macaulay SL, Mitchell CA. Identification of P-Rex1 as a novel Rac1-guanine nucleotide exchange factor (GEF) that promotes actin remodeling and GLUT4 protein trafficking in adipocytes. J Biol Chem 2011; 286:43229-40. [PMID: 22002247 DOI: 10.1074/jbc.m111.306621] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Phosphoinositide 3-kinase (PI3K) signaling promotes the translocation of the glucose transporter, GLUT4, to the plasma membrane in insulin-sensitive tissues to facilitate glucose uptake. In adipocytes, insulin-stimulated reorganization of the actin cytoskeleton has been proposed to play a role in promoting GLUT4 translocation and glucose uptake, in a PI3K-dependent manner. However, the PI3K effectors that promote GLUT4 translocation via regulation of the actin cytoskeleton in adipocytes remain to be fully elucidated. Here we demonstrate that the PI3K-dependent Rac exchange factor, P-Rex1, enhances membrane ruffling in 3T3-L1 adipocytes and promotes GLUT4 trafficking to the plasma membrane at submaximal insulin concentrations. P-Rex1-facilitated GLUT4 trafficking requires a functional actin network and membrane ruffle formation and occurs in a PI3K- and Rac1-dependent manner. In contrast, expression of other Rho GTPases, such as Cdc42 or Rho, did not affect insulin-stimulated P-Rex1-mediated GLUT4 trafficking. P-Rex1 siRNA knockdown or expression of a P-Rex1 dominant negative mutant reduced but did not completely inhibit glucose uptake in response to insulin. Collectively, these studies identify a novel RacGEF in adipocytes as P-Rex1 that, at physiological insulin concentrations, functions as an insulin-dependent regulator of the actin cytoskeleton that contributes to GLUT4 trafficking to the plasma membrane.
Collapse
Affiliation(s)
- Demis Balamatsias
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Chiu TT, Jensen TE, Sylow L, Richter EA, Klip A. Rac1 signalling towards GLUT4/glucose uptake in skeletal muscle. Cell Signal 2011; 23:1546-54. [DOI: 10.1016/j.cellsig.2011.05.022] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Accepted: 05/31/2011] [Indexed: 12/27/2022]
|
18
|
von Lewinski D, Gasser R, Rainer PP, Huber MS, Wilhelm B, Roessl U, Haas T, Wasler A, Grimm M, Bisping E, Pieske B. Functional effects of glucose transporters in human ventricular myocardium. Eur J Heart Fail 2011; 12:106-13. [PMID: 20083620 DOI: 10.1093/eurjhf/hfp191] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Insulin-dependent positive inotropic effects (PIE) are partially Ca(2+) independent. This mechanism is potentially glucose dependent. In contrast to most animal species, human myocardium expresses high levels of sodium-glucose-transporter-1 (SGLT-1) mRNA besides the common glucose-transporters-1 and -4 (GLUT1, GLUT4). METHODS AND RESULTS We used ventricular myocardium from 61 end-stage failing human hearts (ischaemic cardiomyopathy, ICM and dilated cardiomyopathy, DCM) and 13 non-failing donor hearts. The effect of insulin on isometric twitch force was examined with or without blocking of PI3-kinase, GLUT4-translocation, or SGLT-1. Substrate-dependent (glucose vs. pyruvate vs. palmitoyl-carnitine) effects were tested in atrial myocardium. mRNA expression of glucose transporters was analysed. Insulin increased developed force by 122 + or - 7.4, 121.7 + or - 2.5, and 134.1 + or - 5.7% in non-failing, DCM, and ICM (P < 0.05 vs. DCM), respectively. Positive inotropic effect was partially blunted by inhibition of PI-3-kinase, GLUT4, or SGLT1. Combined inhibition of PI3-kinase and glucose-transport completely abolished PIE. Positive inotropic effect was significantly stronger in glucose-containing solution compared with pyruvate or palmitoyl-carnitine containing. mRNA expression showed only a tendency towards elevated GLUT4-expression in ICM. CONCLUSIONS Positive inotropic effect of insulin is pronounced in ICM, but underlying mechanisms are unaltered. The Ca(2+)-independent PIE of insulin is mediated via glucose-transporters. Together with the Ca(2+)-dependent PI-3-kinase mediated pathway, it is responsible for the entire PIE. Substrate-dependency affirms a glucose-dependent part of the PIE.
Collapse
Affiliation(s)
- Dirk von Lewinski
- Department of Cardiology, Working Group Metabolism and Energetics, Medical University Graz, Graz, Austria.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Weems J, Olson AL. Class II histone deacetylases limit GLUT4 gene expression during adipocyte differentiation. J Biol Chem 2010; 286:460-8. [PMID: 21047791 DOI: 10.1074/jbc.m110.157107] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Insulin-dependent glucose homeostasis is highly sensitive to the levels of insulin-responsive glucose transporter 4 (GLUT4) expression in adipocytes. The level of GLUT4 protein expression is highly dependent on the rate of GLUT4 gene transcription. GLUT4 gene transcription is decreased in a variety of physiologic states of insulin resistance including type 2 diabetes, obesity, and prolonged fasting. GLUT4 gene expression in adipocytes is differentiation-dependent, with full expression delayed until late in the differentiation program. In this paper, we have tested the hypothesis that differentiation-dependent GLUT4 gene expression in 3T3-L1 adipocytes is dependent on the nuclear concentration of a class II histone deacetylase (HDAC) protein, HDAC5. We have tested this hypothesis by reducing the levels of class II HDACs in the nuclear compartment of 3T3-L1 preadipocytes using two experimental approaches. First, preadipocytes were treated with phenylephrine, an α-adrenergic receptor agonist, to drive HDACS out of the nuclear compartment. Also, the class II HDAC concentrations were reduced using siRNA knockdown. In each case, reduction of nuclear class II HDAC concentration resulted in increased expression of endogenous GLUT4 mRNA in preadipocytes. Together, our data indicate that class II HDAC expression is the major regulatory mechanism for inhibiting GLUT4 expression in the predifferentiated state.
Collapse
Affiliation(s)
- Juston Weems
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73190, USA
| | | |
Collapse
|
20
|
Chiu TT, Patel N, Shaw AE, Bamburg JR, Klip A. Arp2/3- and cofilin-coordinated actin dynamics is required for insulin-mediated GLUT4 translocation to the surface of muscle cells. Mol Biol Cell 2010; 21:3529-39. [PMID: 20739464 PMCID: PMC2954118 DOI: 10.1091/mbc.e10-04-0316] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Insulin increases GLUT4 at the muscle cell surface, and this process requires actin remodeling. We show that a dynamic cycle of actin polymerization and severing is induced by insulin, governed by Arp2/3 and dephosphorylation of cofilin, respectively. The cycle is self-perpetuating and is essential for GLUT4 translocation. GLUT4 vesicles are actively recruited to the muscle cell surface upon insulin stimulation. Key to this process is Rac-dependent reorganization of filamentous actin beneath the plasma membrane, but the underlying molecular mechanisms have yet to be elucidated. Using L6 rat skeletal myoblasts stably expressing myc-tagged GLUT4, we found that Arp2/3, acting downstream of Rac GTPase, is responsible for the cortical actin polymerization evoked by insulin. siRNA-mediated silencing of either Arp3 or p34 subunits of the Arp2/3 complex abrogated actin remodeling and impaired GLUT4 translocation. Insulin also led to dephosphorylation of the actin-severing protein cofilin on Ser-3, mediated by the phosphatase slingshot. Cofilin dephosphorylation was prevented by strategies depolymerizing remodeled actin (latrunculin B or p34 silencing), suggesting that accumulation of polymerized actin drives severing to enact a dynamic actin cycling. Cofilin knockdown via siRNA caused overwhelming actin polymerization that subsequently inhibited GLUT4 translocation. This inhibition was relieved by reexpressing Xenopus wild-type cofilin-GFP but not the S3E-cofilin-GFP mutant that emulates permanent phosphorylation. Transferrin recycling was not affected by depleting Arp2/3 or cofilin. These results suggest that cofilin dephosphorylation is required for GLUT4 translocation. We propose that Arp2/3 and cofilin coordinate a dynamic cycle of actin branching and severing at the cell cortex, essential for insulin-mediated GLUT4 translocation in muscle cells.
Collapse
Affiliation(s)
- Tim Ting Chiu
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | | | | | | | | |
Collapse
|
21
|
Griesel BA, Weems J, Russell RA, Abel ED, Humphries K, Olson AL. Acute inhibition of fatty acid import inhibits GLUT4 transcription in adipose tissue, but not skeletal or cardiac muscle tissue, partly through liver X receptor (LXR) signaling. Diabetes 2010; 59:800-7. [PMID: 20103707 PMCID: PMC2844827 DOI: 10.2337/db09-1542] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Insulin-mediated glucose uptake is highly sensitive to the levels of the facilitative GLUT protein GLUT4. Transcription of the GLUT4 gene is repressed in states of insulin deficiency and insulin resistance and can be induced by states of enhanced energy output, such as exercise. The cellular signals that regulate GLUT4 transcription are not well understood. We hypothesized that changes in energy substrate flux regulate GLUT4 transcription. RESEARCH DESIGN AND METHODS To test this hypothesis, we used transgenic mice in which expression of the chloramphenicol acetyltransferase (CAT) gene is driven by a functional 895-bp fragment of the human GLUT4 promoter, thereby acting as a reporter for transcriptional activity. Mice were treated with a single dose of etomoxir, which inhibits the transport of long-chain fatty acids into mitochondria and increases basal, but not insulin-mediated, glucose flux. GLUT4 and transgenic CAT mRNA were measured. RESULTS Etomoxir treatment significantly reduced CAT and GLUT4 mRNA transcription in adipose tissue, but did not change transcription in heart and skeletal muscle. Downregulation of GLUT4 transcription was cell autonomous, since etomoxir treatment of 3T3-L1 adipocytes resulted in a similar downregulation of GLUT4 mRNA. GLUT4 transcriptional downregulation required the putative liver X receptor (LXR) binding site in the human GLUT4 gene promoter in adipose tissue and 3T3-L1 adipocytes. Treatment of 3T3-L1 adipocytes with the LXR agonist, TO901317, partially restored GLUT4 expression in etomoxir-treated cells. CONCLUSIONS Our data suggest that long-chain fatty acid import into mitochondria in adipose tissue may produce ligands that regulate expression of metabolic genes.
Collapse
Affiliation(s)
- Beth A. Griesel
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Juston Weems
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Robert A. Russell
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - E. Dale Abel
- Division of Endocrinology, Metabolism, and Diabetes, and Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| | - Kenneth Humphries
- Oklahoma Medical Research Foundation and the Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Ann Louise Olson
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Corresponding author: Ann Louise Olson,
| |
Collapse
|
22
|
Lopez JA, Burchfield JG, Blair DH, Mele K, Ng Y, Vallotton P, James DE, Hughes WE. Identification of a distal GLUT4 trafficking event controlled by actin polymerization. Mol Biol Cell 2009; 20:3918-29. [PMID: 19605560 DOI: 10.1091/mbc.e09-03-0187] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The insulin-stimulated trafficking of GLUT4 to the plasma membrane in muscle and fat tissue constitutes a central process in blood glucose homeostasis. The tethering, docking, and fusion of GLUT4 vesicles with the plasma membrane (PM) represent the most distal steps in this pathway and have been recently shown to be key targets of insulin action. However, it remains unclear how insulin influences these processes to promote the insertion of the glucose transporter into the PM. In this study we have identified a previously uncharacterized role for cortical actin in the distal trafficking of GLUT4. Using high-frequency total internal reflection fluorescence microscopy (TIRFM) imaging, we show that insulin increases actin polymerization near the PM and that disruption of this process inhibited GLUT4 exocytosis. Using TIRFM in combination with probes that could distinguish between vesicle transport and fusion, we found that defective actin remodeling was accompanied by normal insulin-regulated accumulation of GLUT4 vesicles close to the PM, but the final exocytotic fusion step was impaired. These data clearly resolve multiple steps of the final stages of GLUT4 trafficking, demonstrating a crucial role for actin in the final stage of this process.
Collapse
Affiliation(s)
- Jamie A Lopez
- Diabetes and Obesity Research Program, The Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | | | | | | | | | | | | | | |
Collapse
|
23
|
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
|
24
|
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
|
25
|
Garcia-Souza EP, da Silva SV, Félix GB, Rodrigues AL, de Freitas MS, Moura AS, Barja-Fidalgo C. Maternal protein restriction during early lactation induces GLUT4 translocation and mTOR/Akt activation in adipocytes of adult rats. Am J Physiol Endocrinol Metab 2008; 295:E626-36. [PMID: 18559980 DOI: 10.1152/ajpendo.00439.2007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Epidemiological and experimental studies have demonstrated that early postnatal nutrition has been associated with long-term effects on glucose homeostasis in adulthood. Recently, our group demonstrated that undernutrition during early lactation affects the expression and activation of key proteins of the insulin signaling cascade in rat skeletal muscle during postnatal development. To elucidate the molecular mechanisms by which undernutrition during early life leads to changes in insulin sensitivity in peripheral tissues, we investigated the insulin signaling in adipose tissue. Adipocytes were isolated from epididymal fat pads of adult male rats that were the offspring of dams fed either a normal or a protein-free diet during the first 10 days of lactation. The cells were incubated with 100 nM insulin before the assays for immunoblotting analysis, 2-deoxyglucose uptake, immunocytochemistry for GLUT4, and/or actin filaments. Following insulin stimulation, adipocytes isolated from undernourished rats presented reduced tyrosine phosphorylation of IR and IRS-1 and increased basal phosphorylation of IRS-2, Akt, and mTOR compared with controls. Basal glucose uptake was increased in adipocytes from the undernourished group, and the treatment with LY294002 induced only a partial inhibition both in basal and in insulin-stimulated glucose uptake, suggesting an involvement of phosphoinositide 3-kinase activity. These alterations were accompanied by higher GLUT4 content in the plasma membrane and alterations in the actin cytoskeleton dynamics. These data suggest that early postnatal undernutrition impairs insulin sensitivity in adulthood by promoting changes in critical steps of insulin signaling in adipose tissue, which may contribute to permanent changes in glucose homeostasis.
Collapse
Affiliation(s)
- Erica Patrícia Garcia-Souza
- Departament of Pharmacology, Institute of Biology, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, 20551-030, Brasil
| | | | | | | | | | | | | |
Collapse
|
26
|
Insulin action on glucose transporters through molecular switches, tracks and tethers. Biochem J 2008; 413:201-15. [DOI: 10.1042/bj20080723] [Citation(s) in RCA: 214] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Glucose entry into muscle cells is precisely regulated by insulin, through recruitment of GLUT4 (glucose transporter-4) to the membrane of muscle and fat cells. Work done over more than two decades has contributed to mapping the insulin signalling and GLUT4 vesicle trafficking events underpinning this response. In spite of this intensive scientific research, there are outstanding questions that continue to challenge us today. The present review summarizes the knowledge in the field, with emphasis on the latest breakthroughs in insulin signalling at the level of AS160 (Akt substrate of 160 kDa), TBC1D1 (tre-2/USP6, BUB2, cdc16 domain family member 1) and their target Rab proteins; in vesicle trafficking at the level of vesicle mobilization, tethering, docking and fusion with the membrane; and in the participation of the cytoskeleton to achieve optimal temporal and spatial location of insulin-derived signals and GLUT4 vesicles.
Collapse
|
27
|
Sparling DP, Griesel BA, Weems J, Olson AL. GLUT4 enhancer factor (GEF) interacts with MEF2A and HDAC5 to regulate the GLUT4 promoter in adipocytes. J Biol Chem 2008; 283:7429-37. [PMID: 18216015 DOI: 10.1074/jbc.m800481200] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The insulin-responsive glucose transporter, GLUT4, is regulated in various physiologic states at the transcriptional level. When expressed in transgenic mice, the human GLUT4 promoter is governed by two cis-acting sequences: an MEF2 binding domain and Domain I, that function both as positive and negative regulators depending on the physiologic state. MEF2 proteins and GLUT4 enhancer factor (GEF) are known ligands for these cis-acting elements, but their mechanism of action is unclear. To begin to understand this important process, we have characterized GEF structural domains and its interactions with the MEF2A isoform. We find that the C terminus of GEF comprises its DNA-binding domain, but does not contribute to GEF homo-oligomerization. We also have found that GEF dimerizes with increased affinity to a hypophosphorylated form of MEF2A. Furthermore, we demonstrated that MEF2A binding to its cognate binding site can increase the DNA binding activity of GEF to Domain I, suggesting a novel mechanism for MEF2A transcriptional activation. Finally, we have demonstrated that the transcriptional co-repressor HDAC5 can interact with GEF in the absence of MEF2 proteins and specifically inhibit GLUT4 promoter activity. These findings lead to the hypothesis that GEF and the MEF2 proteins form a complex on the GLUT4 promoter that allows for recruitment of transcriptional co-regulators (repressors and/or activators) to control GLUT4 promoter activity.
Collapse
Affiliation(s)
- David P Sparling
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73190, USA
| | | | | | | |
Collapse
|
28
|
Brown AE, Yeaman SJ, Walker M. Targeted suppression of calpain-10 expression impairs insulin-stimulated glucose uptake in cultured primary human skeletal muscle cells. Mol Genet Metab 2007; 91:318-24. [PMID: 17572128 DOI: 10.1016/j.ymgme.2007.05.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2007] [Revised: 05/01/2007] [Accepted: 05/02/2007] [Indexed: 12/27/2022]
Abstract
Calpain-10 was identified as a novel type 2 diabetes susceptibility gene, although the mechanisms by which it increases susceptibility to type 2 diabetes remain unclear. As skeletal muscle is the principal site of the peripheral insulin resistance for glucose disposal in type 2 diabetes, we investigated whether targeted suppression of calpain-10 expression directly affects insulin action in cultured human skeletal muscle cells. Short interfering RNAs (siRNAs) were employed to specifically suppress CAPN10 gene expression. Suppression was seen at both the transcript and protein level, as assessed by quantitative PCR and Western blotting. Suppression of CAPN10 mRNA expression (75% decrease compared to untransfected myotubes) was associated with a significant decrease (p=0.04) in insulin-stimulated glucose uptake (1.03+/-0.06 [mean+/-SEM]-fold increase over basal) compared to the untransfected myotubes (1.43+/-0.16-fold increase). In contrast, decreased suppression of calpain-10 expression did not affect insulin-stimulated glycogen synthesis nor insulin-stimulated phosphorylation of protein kinase B, a key component of the insulin-signalling pathway. This study confirms that calpain-10 plays a role in insulin-stimulated glucose uptake in human skeletal muscle cells. Suppression of calpain-10 expression did not affect insulin-stimulated glycogen synthesis nor insulin-signalling via PKB, suggesting that calpain-10 may exert a direct regulatory effect upon the glucose uptake mechanism.
Collapse
Affiliation(s)
- Audrey E Brown
- School of Clinical Medical Sciences, University of Newcastle upon Tyne, Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | | | | |
Collapse
|
29
|
Yang C, Yang Y, Gupta N, Liu X, He A, Liu L, Zuo J, Chang Y, Fang F. Pentaspan membrane glycoprotein, prominin-1, is involved in glucose metabolism and cytoskeleton alteration. BIOCHEMISTRY (MOSCOW) 2007; 72:854-62. [DOI: 10.1134/s000629790708007x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
30
|
Liu XJ, Yang C, Gupta N, Zuo J, Chang YS, Fang FD. Protein kinase C-zeta regulation of GLUT4 translocation through actin remodeling in CHO cells. J Mol Med (Berl) 2007; 85:851-61. [PMID: 17619838 DOI: 10.1007/s00109-007-0232-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2007] [Revised: 04/25/2007] [Accepted: 05/31/2007] [Indexed: 01/08/2023]
Abstract
Actin remodeling plays a crucial role in insulin-induced translocation of glucose transporter 4 (GLUT4) from the cytoplasm to the plasma membrane and subsequent glucose transport. Protein kinase C (PKC) zeta has been implicated in this translocation process, although the exact mechanism remains unknown. In this study, we investigated the effect of PKCzeta on actin cytoskeleton and translocation of GLUT4 in CHO-K1 cells expressing myc-tagged GLUT4. Insulin stimulated the phosphorylation of PKCzeta at Thr410 with no apparent effect on its protein expression. Moreover, insulin promoted colocalization of PKCzeta and actin that could be abolished by Latrunculin B. The overexpression of PKCzeta mimicked the insulin-induced change in actin cytoskeleton and translocation of GLUT4. These effects were also completely abrogated by Latrunculin B treatment. Using cell-permeable pseudosubstrate (PS) inhibitor of PKCzeta, the response to insulin could be alleviated. Our results strongly suggest that PKCzeta mediates the stimulatory effect of insulin on GLUT4 translocation through its interaction with actin cytoskeleton.
Collapse
Affiliation(s)
- Xiao-Jun Liu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, China
| | | | | | | | | | | |
Collapse
|
31
|
Funaki M, Benincasa K, Randhawa PK. Peptide rescues GLUT4 recruitment, but not GLUT4 activation, in insulin resistance. Biochem Biophys Res Commun 2007; 360:891-6. [PMID: 17631270 DOI: 10.1016/j.bbrc.2007.06.153] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2007] [Accepted: 06/29/2007] [Indexed: 11/30/2022]
Abstract
Insulin-stimulated GLUT4 recruitment to the plasma membrane is impaired in insulin resistance. We recently reported that a cell permeable phosphoinositide-binding peptide induces GLUT4 recruitment as potently as insulin, but does not activate GLUT4 to initiate glucose uptake. Here we investigated whether the peptide-induced GLUT4 recruitment is intact in insulin resistance. The expression levels of GLUT1 and GLUT4 were unaffected by chronically treating 3T3-L1 adipocytes with insulin. GLUT4 recruitment by acute insulin stimulation after chronic insulin treatment was significantly reduced, but was fully restored by the peptide treatment. However, subsequent acute insulin stimulation to activate GLUT4 failed to increase glucose uptake in peptide-pretreated cells. Insulin-stimulated GLUT1 recruitment was unaffected by the peptide pretreatment. These results suggest that the GLUT4 recruitment signal caused by the peptide is intact in insulin resistance, but GLUT4 activation that occurs subsequent to recruitment is not rescued by the peptide treatment.
Collapse
Affiliation(s)
- Makoto Funaki
- Department of Physiology, Institute for Medicine & Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | | | |
Collapse
|
32
|
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
|
33
|
Muretta JM, Romenskaia I, Cassiday PA, Mastick CC. Expression of a synapsin IIb site 1 phosphorylation mutant in 3T3-L1 adipocytes inhibits basal intracellular retention of Glut4. J Cell Sci 2007; 120:1168-77. [PMID: 17341582 DOI: 10.1242/jcs.03413] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Glut4 exocytosis in adipocytes uses protein machinery that is shared with other regulated secretory processes. Synapsins are phosphoproteins that regulate a ;reserve pool' of vesicles clustered behind the active zone in neurons. We found that adipocytes (primary cells and the 3T3-L1 cell line) express synapsin IIb mRNA and protein. Synapsin IIb co-localizes with Glut4 in perinuclear vesicle clusters. To test whether synapsin plays a role in Glut4 traffic, a site 1 phosphorylation mutant (S10A synapsin) was expressed in 3T3-L1 adipocytes. Interestingly, expression of S10A synapsin increased basal cell surface Glut4 almost fourfold (50% maximal insulin effect). Insulin caused a further twofold translocation of Glut4 in these cells. Expression of the N-terminus of S10A synapsin (amino acids 1-118) was sufficient to inhibit basal Glut4 retention. Neither wild-type nor S10D synapsin redistributed Glut4. S10A synapsin did not elevate surface levels of the transferrin receptor in adipocytes or Glut4 in fibroblasts. Therefore, S10A synapsin is inhibiting the specialized process of basal intracellular retention of Glut4 in adipocytes, without affecting general endocytic cycling. While mutant forms of many proteins inhibit Glut4 exocytosis in response to insulin, S10A synapsin is one of only a few that specifically inhibits Glut4 retention in basal adipocytes. These data indicate that the synapsins are important regulators of membrane traffic in many cell types.
Collapse
Affiliation(s)
- Joseph M Muretta
- Department of Biochemistry and Molecular Biology, Mailstop 330, University of Nevada, Reno, NV 89557, USA
| | | | | | | |
Collapse
|
34
|
Gururajan M, Dasu T, Shahidain S, Jennings CD, Robertson DA, Rangnekar VM, Bondada S. Spleen tyrosine kinase (Syk), a novel target of curcumin, is required for B lymphoma growth. THE JOURNAL OF IMMUNOLOGY 2007; 178:111-21. [PMID: 17182546 DOI: 10.4049/jimmunol.178.1.111] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Curcumin (diferuloylmethane), a component of dietary spice turmeric (Curcuma longa), has been shown in recent studies to have therapeutic potential in the treatment of cancer, diabetes, arthritis, and osteoporosis. We investigated the ability of curcumin to modulate the growth of B lymphomas. Curcumin inhibited the growth of both murine and human B lymphoma in vitro and murine B lymphoma in vivo. We also demonstrate that curcumin-mediated growth inhibition of B lymphoma is through inhibition of the survival kinase Akt and its key target Bad. However, in vitro kinase assays show that Akt is not a direct target of curcumin. We identified a novel target for curcumin in B lymphoma viz spleen tyrosine kinase (Syk). Syk is constitutively activated in primary tumors and B lymphoma cell lines and curcumin down-modulates Syk activity accompanied by down-regulation of Akt activation. Moreover, we show that overexpression of Akt, a target of Syk, or Bcl-x(L), a target of Akt can overcome curcumin-induced apoptosis of B lymphoma cells. These observations suggest a novel growth promoting role for Syk in lymphoma cells.
Collapse
Affiliation(s)
- Murali Gururajan
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
JeBailey L, Wanono O, Niu W, Roessler J, Rudich A, Klip A. Ceramide- and oxidant-induced insulin resistance involve loss of insulin-dependent Rac-activation and actin remodeling in muscle cells. Diabetes 2007; 56:394-403. [PMID: 17259384 DOI: 10.2337/db06-0823] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In muscle cells, insulin elicits recruitment of the glucose transporter GLUT4 to the plasma membrane. This process engages sequential signaling from insulin receptor substrate (IRS)-1 to phosphatidylinositol (PI) 3-kinase and the serine/threonine kinase Akt. GLUT4 translocation also requires an Akt-independent but PI 3-kinase-and Rac-dependent remodeling of filamentous actin. Although IRS-1 phosphorylation is often reduced in insulin-resistant states in vivo, several conditions eliciting insulin resistance in cell culture spare this early step. Here, we show that insulin-dependent Rac activation and its consequent actin remodeling were abolished upon exposure of L6 myotubes beginning at doses of C2-ceramide or oxidant-producing glucose oxidase as low as 12.5 micromol/l and 12.5 mU/ml, respectively. At 25 micromol/l and 25 mU/ml, glucose oxidase and C2-ceramide markedly reduced GLUT4 translocation and glucose uptake and lowered Akt phosphorylation on Ser473 and Thr308, yet they affected neither IRS-1 tyrosine phosphorylation nor its association with p85 and PI 3-kinase activity. Small interfering RNA-dependent Rac1 knockdown prevented actin remodeling and GLUT4 translocation but spared Akt phosphorylation, suggesting that Rac and actin remodeling do not contribute to overall Akt activation. We propose that ceramide and oxidative stress can each affect two independent arms of insulin signaling to GLUT4 at distinct steps, Rac-GTP loading and Akt phosphorylation.
Collapse
Affiliation(s)
- Lellean JeBailey
- Programme in Cell Biology, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, Canada M5G 1X8
| | | | | | | | | | | |
Collapse
|
36
|
Eyster CA, Duggins QS, Gorbsky GJ, Olson AL. Microtubule network is required for insulin signaling through activation of Akt/protein kinase B: evidence that insulin stimulates vesicle docking/fusion but not intracellular mobility. J Biol Chem 2006; 281:39719-27. [PMID: 17068336 DOI: 10.1074/jbc.m607101200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The microtubule network has been shown to be required for insulin-dependent GLUT4 redistribution; however, the precise molecular function has not been elucidated. In this article, we used fluorescence recovery after photobleaching (FRAP) to evaluate the role of microtubules in intracellular GLUT4 vesicle mobility. A comparison of the rate of fluorescence recovery (t((1/2))), and the maximum fluorescence recovered (F(max)) was made between basal and insulin-treated cells with or without nocodazole treatment to disrupt microtubules. We found that intracellular mobility of fluorescently tagged GLUT4 (HA-GLUT4-GFP) was high in basal cells. Mobility was not increased by insulin treatment. Basal mobility was dependent upon an intact microtubule network. Using a constitutively active Akt to signal GLUT4 redistribution, we found that microtubule-based GLUT4 vesicle mobility was not obligatory for GLUT4 plasma membrane insertion. Our findings suggest that microtubules organize the insulin-signaling complex and provide a surface for basal mobility of GLUT4 vesicles. Our data do not support an obligatory requirement for long range microtubule-based movement of GLUT4 vesicles for insulin-mediated GLUT4 redistribution to the cell surface. Taken together, these findings suggest a model in which insulin signaling targets membrane docking and/or fusion rather than GLUT4 trafficking to the cell surface.
Collapse
Affiliation(s)
- Craig A Eyster
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | | | | | | |
Collapse
|
37
|
Shikano S, Coblitz B, Wu M, Li M. 14-3-3 proteins: regulation of endoplasmic reticulum localization and surface expression of membrane proteins. Trends Cell Biol 2006; 16:370-5. [PMID: 16769213 DOI: 10.1016/j.tcb.2006.05.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2006] [Revised: 03/31/2006] [Accepted: 05/24/2006] [Indexed: 11/24/2022]
Abstract
The density and composition of cell surface proteins are major determinants for cellular functions. Regulation of cell surface molecules occurs at several levels, including the efficiency of surface transport, and is therefore of great interest. As the major phosphoprotein-binding modules, 14-3-3 proteins are known for their crucial roles in a wide range of cellular activities, including the subcellular localization of target proteins. Accumulating evidence suggests a role for 14-3-3 in surface transport of membrane proteins, in which 14-3-3 binding reduces endoplasmic reticulum (ER) localization, thereby promoting surface expression of membrane proteins. Here, we focus on recent evidence of 14-3-3-mediated surface transport and discuss the possible molecular mechanisms.
Collapse
Affiliation(s)
- Sojin Shikano
- Department of Neuroscience and High Throughput Biology Center, School of Medicine, Johns Hopkins University, 733 North Broadway, Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
38
|
Liu L, Jedrychowski MP, Gygi SP, Pilch PF. Role of insulin-dependent cortical fodrin/spectrin remodeling in glucose transporter 4 translocation in rat adipocytes. Mol Biol Cell 2006; 17:4249-56. [PMID: 16870704 PMCID: PMC1635356 DOI: 10.1091/mbc.e06-04-0278] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Fodrin or nonerythroid spectrin is an abundant component of the cortical cytoskeletal network in rat adipocytes. Fodrin has a highly punctate distribution in resting cells, and insulin causes a dramatic remodeling of fodrin to a more diffuse pattern. Insulin-mediated remodeling of actin occurs to a lesser extent than does that of fodrin. We show that fodrin interacts with the t-soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) syntaxin 4, and this interaction is increased by insulin stimulation and decreased by prior latrunculin A treatment. Latrunculin A disrupts all actin filaments, inhibits glucose transporter 4 (GLUT4) translocation, and causes fodrin to partially redistribute from the plasma membrane to the cytosol. In contrast, cytochalasin D disrupts only the short actin filament signal, and cytochalasin D neither inhibits GLUT4 translocation nor fodrin redistribution in adipocytes. Together, our data suggest that insulin induces remodeling of the fodrin-actin network, which is required for the fusion of GLUT4 storage vesicles with the plasma membrane by permitting their access to the t-SNARE syntaxin 4.
Collapse
Affiliation(s)
- Libin Liu
- *Department of Biochemistry, Boston University Medical School, Boston, MA 02118; and
| | - Mark P. Jedrychowski
- *Department of Biochemistry, Boston University Medical School, Boston, MA 02118; and
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | - Paul F. Pilch
- *Department of Biochemistry, Boston University Medical School, Boston, MA 02118; and
| |
Collapse
|
39
|
Funaki M, DiFransico L, Janmey PA. PI 4,5-P2 stimulates glucose transport activity of GLUT4 in the plasma membrane of 3T3-L1 adipocytes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2006; 1763:889-99. [PMID: 16828894 PMCID: PMC3118463 DOI: 10.1016/j.bbamcr.2006.05.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2006] [Revised: 05/08/2006] [Accepted: 05/09/2006] [Indexed: 12/16/2022]
Abstract
Insulin-stimulated glucose uptake through GLUT4 plays a pivotal role in maintaining normal blood glucose levels. Glucose transport through GLUT4 requires both GLUT4 translocation to the plasma membrane and GLUT4 activation at the plasma membrane. Here we report that a cell-permeable phosphoinositide-binding peptide, which induces GLUT4 translocation without activation, sequestered PI 4,5-P2 in the plasma membrane from its binding partners. Restoring PI 4,5-P2 to the plasma membrane after the peptide treatment increased glucose uptake. No additional glucose transporters were recruited to the plasma membrane, suggesting that the increased glucose uptake was attributable to GLUT4 activation. Cells overexpressing phosphatidylinositol-4-phosphate 5-kinase treated with the peptide followed by its removal exhibited a higher level of glucose transport than cells stimulated with a submaximal level of insulin. However, only cells treated with submaximal insulin exhibited translocation of the PH-domains of the general receptor for phosphoinositides (GRP1) to the plasma membrane. Thus, PI 4,5-P2, but not PI 3,4,5-P3 converted from PI 4,5-P2, induced GLUT4 activation. Inhibiting F-actin remodeling after the peptide treatment significantly impaired GLUT4 activation induced either by PI 4,5-P2 or by insulin. These results suggest that PI 4,5-P2 in the plasma membrane acts as a second messenger to activate GLUT4, possibly through F-actin remodeling.
Collapse
Affiliation(s)
- Makoto Funaki
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, 1080 Vagelos Research Laboratories, 3340 Smith Walk, Philadelphia, 19104, USA.
| | | | | |
Collapse
|
40
|
Bedi D, Clarke KJ, Dennis JC, Zhong Q, Brunson BL, Morrison EE, Judd RL. Endothelin-1 inhibits adiponectin secretion through a phosphatidylinositol 4,5-bisphosphate/actin-dependent mechanism. Biochem Biophys Res Commun 2006; 345:332-9. [PMID: 16682005 DOI: 10.1016/j.bbrc.2006.04.098] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2006] [Accepted: 04/19/2006] [Indexed: 11/28/2022]
Abstract
Adiponectin is an adipokine with profound insulin-sensitizing, anti-inflammatory, and anti-atherogenic properties. Plasma levels of adiponectin are reduced in insulin resistant states such as obesity, type 2 diabetes and cardiovascular disease. However, the mechanism(s) by which adiponectin concentrations are decreased during disease development is unclear. Studies have shown that endothelin-1 (ET-1), a vasoconstrictor peptide, affects adipocyte glucose metabolism and secretion of adipokines such as leptin, resistin, and adiponectin. The goal of our study was to determine the mechanism by which ET-1 decreases adiponectin secretion. 3T3-L1 adipocytes were treated for 24h with ET-1 (10nM) and then stimulated with vehicle or insulin (100 nM) for a period of 1-2h. Chronic ET-1 (24h) treatment significantly decreased basal and insulin-stimulated adiponectin secretion by 66% and 47%, respectively. Inhibition of phosphatidylinositol 4,5-bisphosphate (PIP(2)) hydrolysis by the PLCbeta inhibitor, U73122, or exogenous addition of PIP(2):histone carrier complex (1.25:0.625 microM) ameliorated the decrease in basal and insulin-stimulated adiponectin secretion observed with ET-1. However, treatment with exogenous PIP(2):histone carrier complex and the actin depolymerizing agent latrunculin B (20 microM) did not reverse the ET-1-mediated decrease in adiponectin secretion. In conclusion, we demonstrate that ET-1 inhibits basal and insulin-stimulated adiponectin secretion through PIP(2) modulation of the actin cytoskeleton.
Collapse
Affiliation(s)
- Deepa Bedi
- Boshell Diabetes and Metabolic Diseases Research Program, Department of Anatomy, Physiology and Pharmacology, Auburn University, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Thomas EC, Zhe Y, Molero JC, Schmitz-Peiffer C, Ramm G, James DE, Whitehead JP. The subcellular fractionation properties and function of insulin receptor substrate-1 (IRS-1) are independent of cytoskeletal integrity. Int J Biochem Cell Biol 2006; 38:1686-99. [PMID: 16702017 DOI: 10.1016/j.biocel.2006.03.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2006] [Revised: 03/15/2006] [Accepted: 03/20/2006] [Indexed: 10/24/2022]
Abstract
Efficient insulin action requires spatial and temporal coordination of signaling cascades. The prototypical insulin receptor substrate, IRS-1 plays a central role in insulin signaling. By subcellular fractionation IRS-1 is enriched in a particulate fraction, termed the high speed pellet (HSP), and its redistribution from this fraction is associated with signal attenuation and insulin resistance. Anecdotal evidence suggests the cytoskeleton may underpin the localization of IRS-1 to the HSP. In the present study we have taken a systematic approach to examine whether the cytoskeleton contributes to the subcellular fractionation properties and function of IRS-1. By standard microscopy or immunoprecipitation we were unable to detect evidence to support a specific interaction between IRS-1 and the major cytoskeletal components actin (microfilaments), vimentin (intermediate filaments), and tubulin (microtubules) in 3T3-L1 adipocytes or in CHO.IR.IRS-1 cells. Pharmacological disruption of microfilaments and microtubules, individually or in combination, was without effect on the subcellular distribution of IRS-1 or insulin-stimulated tyrosine phosphorylation in either cell type. Phosphorylation of Akt was modestly reduced (20-35%) in 3T3-L1 adipocytes but not in CHO.IR.IRS-1 cells. In cells lacking intermediate filaments (Vim(-/-)) IRS-1 expression, distribution and insulin-stimulated phosphorylation appeared normal. Even after depolymerisation of microfilaments and microtubules, insulin-stimulated phosphorylation of IRS-1 and Akt were maintained in Vim(-/-) cells. Taken together these data indicate that the characteristic subcellular fractionation properties and function of IRS-1 are unlikely to be mediated by cytoskeletal networks and that proximal insulin signaling does not require an intact cytoskeleton.
Collapse
Affiliation(s)
- Elaine C Thomas
- Centre for Diabetes and Endocrine Research, Princess Alexandra Hospital, University of Queensland, Brisbane, Qld 4102, Australia
| | | | | | | | | | | | | |
Collapse
|
42
|
Milsom C, Rak J. Regulation of tissue factor and angiogenesis-related genes by changes in cell shape. Biochem Biophys Res Commun 2005; 337:1267-75. [PMID: 16236262 DOI: 10.1016/j.bbrc.2005.09.187] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2005] [Accepted: 09/29/2005] [Indexed: 11/30/2022]
Abstract
During development, tissue injury, and cancer, epithelial cells engage in communication with the vascular system by using several molecular mediators acting directly or through changes in the haemostatic system.The latter category is epitomised by the procoagulant cellular receptor known as tissue factor (TF). Here, we show that when cellular architecture is altered by a shift in culture conditions from monolayer to three-dimensional multicellular spheroids, expression of multiple angiogenesis effectors (VEGF, TSP-1, TSP-2, Ang-1, and TF) is profoundly altered. In particular, TF is dramatically upregulated in a transformed murine breast epithelial cell line (EMT6) under these conditions. This appears to be linked to a particular change in cell shape and cytoskeletal (actin) reorganisation, as treatment of these cells with cytochalasin D (Cyt D), but not with latrunculin B, recapitulates and potentiates TF upregulation. Collectively, these results suggest that the ability of epithelial cells to interact with the vascular system via expression of the TF gene (and other effectors) is under the control of complex alterations in cellular architecture.
Collapse
Affiliation(s)
- Chloe Milsom
- Henderson Research Centre, McMaster University, Hamilton, Ont., Canada L8V 1C3
| | | |
Collapse
|
43
|
Patel N, Huang C, Klip A. Cellular location of insulin-triggered signals and implications for glucose uptake. Pflugers Arch 2005; 451:499-510. [PMID: 16284741 DOI: 10.1007/s00424-005-1475-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2005] [Accepted: 06/02/2005] [Indexed: 12/29/2022]
Abstract
Insulin stimulation of glucose uptake into muscle and fat cells requires movement of GLUT4-containing vesicles from intracellular compartments to the plasma membrane. Accordingly, insulin-derived signals must arrive at and be recognized by the appropriate intracellular GLUT4 pools. We describe the insulin signals participating in GLUT4 translocation, and review evidence that they are recruited to intracellular membranes in conjunction with cytoskeletal elements. Such segregation may facilitate the encounter between signals and target vesicles. In most animal and cellular models of insulin resistance, insulin-stimulated GLUT4 translocation to the plasma membrane is reduced. Insulin resistance caused by oxidative stress does not affect early insulin signals, rather their intracellular localization is altered. In this and several other insulin-resistant states, insulin-induced actin remodelling is concomitantly diminished. We summarize evidence suggesting that spatial localization of signals is critical for efficient insulin action, and that the cytoskeleton may act as a scaffold to promote efficient translocation of GLUT4 to the cell surface.
Collapse
Affiliation(s)
- Nish Patel
- Programme in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | |
Collapse
|
44
|
Thong FSL, Dugani CB, Klip A. Turning signals on and off: GLUT4 traffic in the insulin-signaling highway. Physiology (Bethesda) 2005; 20:271-84. [PMID: 16024515 DOI: 10.1152/physiol.00017.2005] [Citation(s) in RCA: 161] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Insulin stimulation of glucose uptake into skeletal muscle and adipose tissues is achieved by accelerating glucose transporter GLUT4 exocytosis from intracellular compartments to the plasma membrane and minimally reducing its endocytosis. The round trip of GLUT4 is intricately regulated by diverse signaling molecules impinging on specific compartments. Here we highlight the key molecular signals that are turned on and off by insulin to accomplish this task.
Collapse
Affiliation(s)
- Farah S L Thong
- Programme in Cell Biology, The Hospital for Sick Children, Ontario, Canada
| | | | | |
Collapse
|