1
|
Wang Q, Peng W, Yang Y, Wu Y, Han R, Ding T, Zhang X, Liu J, Yang J, Liu J. Proteome and ubiquitinome analyses of the brain cortex in K18- hACE2 mice infected with SARS-CoV-2. iScience 2024; 27:110602. [PMID: 39211577 PMCID: PMC11357812 DOI: 10.1016/j.isci.2024.110602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/03/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
Clinical research indicates that SARS-CoV-2 infection is linked to several neurological consequences, and the virus is still spreading despite the availability of vaccinations and antiviral medications. To determine how hosts respond to SARS-CoV-2 infection, we employed LC-MS/MS to perform ubiquitinome and proteome analyses of the brain cortexes from K18-hACE2 mice in the presence and absence of SARS-CoV-2 infection. A total of 8,024 quantifiable proteins and 5,220 quantifiable lysine ubiquitination (Kub) sites in 2023 proteins were found. Glutamatergic synapse, calcium signaling pathway, and long-term potentiation may all play roles in the neurological consequences of SARS-CoV-2 infection. Then, we observed possible interactions between 26 SARS-CoV-2 proteins/E3 ubiquitin-protein ligases/deubiquitinases and several differentially expressed mouse proteins or Kub sites. We present the first description of the brain cortex ubiquitinome in K18-hACE2 mice, laying the groundwork for further investigation into the pathogenic processes and treatment options for neurological dysfunction following SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Qiaochu Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Wanjun Peng
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China
| | - Yehong Yang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Yue Wu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Rong Han
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Tao Ding
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Xutong Zhang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Jiangning Liu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, CAMS and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China
| | - Juntao Yang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Jiangfeng Liu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
2
|
Looser ZJ, Faik Z, Ravotto L, Zanker HS, Jung RB, Werner HB, Ruhwedel T, Möbius W, Bergles DE, Barros LF, Nave KA, Weber B, Saab AS. Oligodendrocyte-axon metabolic coupling is mediated by extracellular K + and maintains axonal health. Nat Neurosci 2024; 27:433-448. [PMID: 38267524 PMCID: PMC10917689 DOI: 10.1038/s41593-023-01558-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/13/2023] [Indexed: 01/26/2024]
Abstract
The integrity of myelinated axons relies on homeostatic support from oligodendrocytes (OLs). To determine how OLs detect axonal spiking and how rapid axon-OL metabolic coupling is regulated in the white matter, we studied activity-dependent calcium (Ca2+) and metabolite fluxes in the mouse optic nerve. We show that fast axonal spiking triggers Ca2+ signaling and glycolysis in OLs. OLs detect axonal activity through increases in extracellular potassium (K+) concentrations and activation of Kir4.1 channels, thereby regulating metabolite supply to axons. Both pharmacological inhibition and OL-specific inactivation of Kir4.1 reduce the activity-induced axonal lactate surge. Mice lacking oligodendroglial Kir4.1 exhibit lower resting lactate levels and altered glucose metabolism in axons. These early deficits in axonal energy metabolism are associated with late-onset axonopathy. Our findings reveal that OLs detect fast axonal spiking through K+ signaling, making acute metabolic coupling possible and adjusting the axon-OL metabolic unit to promote axonal health.
Collapse
Affiliation(s)
- Zoe J Looser
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Zainab Faik
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Luca Ravotto
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Henri S Zanker
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Ramona B Jung
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Torben Ruhwedel
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Dwight E Bergles
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - L Felipe Barros
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Aiman S Saab
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
3
|
Diaz-Vegas A, Norris DM, Jall-Rogg S, Cooke KC, Conway OJ, Shun-Shion AS, Duan X, Potter M, van Gerwen J, Baird HJ, Humphrey SJ, James DE, Fazakerley DJ, Burchfield JG. A high-content endogenous GLUT4 trafficking assay reveals new aspects of adipocyte biology. Life Sci Alliance 2023; 6:e202201585. [PMID: 36283703 PMCID: PMC9595207 DOI: 10.26508/lsa.202201585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/10/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022] Open
Abstract
Insulin-induced GLUT4 translocation to the plasma membrane in muscle and adipocytes is crucial for whole-body glucose homeostasis. Currently, GLUT4 trafficking assays rely on overexpression of tagged GLUT4. Here we describe a high-content imaging platform for studying endogenous GLUT4 translocation in intact adipocytes. This method enables high fidelity analysis of GLUT4 responses to specific perturbations, multiplexing of other trafficking proteins and other features including lipid droplet morphology. Using this multiplexed approach we showed that Vps45 and Rab14 are selective regulators of GLUT4, but Trarg1, Stx6, Stx16, Tbc1d4 and Rab10 knockdown affected both GLUT4 and TfR translocation. Thus, GLUT4 and TfR translocation machinery likely have some overlap upon insulin-stimulation. In addition, we identified Kif13A, a Rab10 binding molecular motor, as a novel regulator of GLUT4 traffic. Finally, comparison of endogenous to overexpressed GLUT4 highlights that the endogenous GLUT4 methodology has an enhanced sensitivity to genetic perturbations and emphasises the advantage of studying endogenous protein trafficking for drug discovery and genetic analysis of insulin action in relevant cell types.
Collapse
Affiliation(s)
- Alexis Diaz-Vegas
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, Australia
| | - Dougall M Norris
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Sigrid Jall-Rogg
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, Australia
| | - Kristen C Cooke
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, Australia
| | - Olivia J Conway
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Amber S Shun-Shion
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Xiaowen Duan
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Meg Potter
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, Australia
| | - Julian van Gerwen
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, Australia
| | - Harry Jm Baird
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Sean J Humphrey
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, Australia
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, Australia
- School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Daniel J Fazakerley
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - James G Burchfield
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, Australia
| |
Collapse
|
4
|
Liu SJ, Cai TH, Fang CL, Lin SZ, Yang WQ, Wei Y, Zhou F, Liu L, Luo Y, Guo ZY, Zhao G, Li YP, Li LM. Long-term exercise training down-regulates m 6A RNA demethylase FTO expression in the hippocampus and hypothalamus: an effective intervention for epigenetic modification. BMC Neurosci 2022; 23:54. [PMID: 36163017 PMCID: PMC9513931 DOI: 10.1186/s12868-022-00742-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/15/2022] [Indexed: 11/14/2022] Open
Abstract
Background Exercise boosts the health of some brain parts, such as the hippocampus and hypothalamus. Several studies show that long-term exercise improves spatial learning and memory, enhances hypothalamic leptin sensitivity, and regulates energy balance. However, the effect of exercise on the hippocampus and hypothalamus is not fully understood. The study aimed to find epigenetic modifications or changes in gene expression of the hippocampus and hypothalamus due to exercise. Methods Male C57BL/6 mice were randomly divided into sedentary and exercise groups. All mice in the exercise group were subjected to treadmill exercise 5 days per week for 1 h each day. After the 12-week exercise intervention, the hippocampus and hypothalamus tissue were used for RNA-sequencing or molecular biology experiments. Results In both groups, numerous differentially expressed genes of the hippocampus (up-regulated: 53, down-regulated: 49) and hypothalamus (up-regulated: 24, down-regulated: 40) were observed. In the exercise group, increased level of N6-methyladenosine (m6A) was observed in the hippocampus and hypothalamus (p < 0.05). Furthermore, the fat mass and obesity-associated gene (FTO) of the hippocampus and hypothalamus were down-regulated in the exercise group (p < 0.001). In addition, the Fto co-expression genes of the mouse brain were studied and analyzed using database to determine the potential roles of exercise-downregulated FTO in the brain. Conclusion The findings demonstrate that long-term exercise might elevates the levels of m6A-tagged transcripts in the hippocampus and hypothalamus via down-regulation of FTO. Hence, exercise might be an effective intervention for epigenetic modification.
Collapse
Affiliation(s)
- Shu-Jing Liu
- Center for Scientific Research and Institute of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Tong-Hui Cai
- The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Chun-Lu Fang
- Center for Scientific Research and Institute of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Shao-Zhang Lin
- The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Wen-Qi Yang
- Center for Scientific Research and Institute of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Yuan Wei
- Center for Scientific Research and Institute of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Fu Zhou
- Center for Scientific Research and Institute of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Ling Liu
- Center for Scientific Research and Institute of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Yuan Luo
- Center for Scientific Research and Institute of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Zi-Yi Guo
- Center for Scientific Research and Institute of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Ge Zhao
- Center for Scientific Research and Institute of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Ya-Ping Li
- Center for Scientific Research and Institute of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Liang-Ming Li
- Center for Scientific Research and Institute of Exercise and Health, Guangzhou Sport University, Guangzhou, China.
| |
Collapse
|
5
|
Kang BB, Chiang BH. A novel phenolic formulation for treating hepatic and peripheral insulin resistance by regulating GLUT4-mediated glucose uptake. J Tradit Complement Med 2022; 12:195-205. [PMID: 35528476 PMCID: PMC9072824 DOI: 10.1016/j.jtcme.2021.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/07/2021] [Accepted: 08/07/2021] [Indexed: 11/26/2022] Open
|
6
|
Huang X, Yang J, Huang X, Zhang Z, Liu J, Zou L, Yang X. Tetramethylpyrazine Improves Cognitive Impairment and Modifies the Hippocampal Proteome in Two Mouse Models of Alzheimer's Disease. Front Cell Dev Biol 2021; 9:632843. [PMID: 33791294 PMCID: PMC8005584 DOI: 10.3389/fcell.2021.632843] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/15/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD), one of the most common neurodegenerative diseases, has no effective treatment. We studied the potential effects of tetramethylpyrazine (TMP), an alkaloid in the rhizome of Ligusticum chuanxiong Hort. used in Traditional Chinese Medicine (chuānxiong) to treat ischemic stroke, on AD progression in two AD mouse models. Eight-month-old 3xTg-AD mice received TMP treatment (10 mg/kg/d) for 1 month, and 4-month-old APP/PS1-AD mice received TMP treatment (10 mg/kg/d) for 2 months. Behavioral tests, including step-down passive avoidance (SDA), new object recognition (NOR), Morris water maze (MWM), and Contextual fear conditioning test showed that TMP significantly improved the learning and memory of the two AD-transgenic mice. In addition, TMP reduced beta-amyloid (Aß) levels and tau phosphorylation (p-tau). Venny map pointed out that 116 proteins were commonly changed in 3xTg mice vs. wild type (WT) mice and TMP-treated mice vs. -untreated mice. The same 130 proteins were commonly changed in APP/PS1 mice vs. WT mice and TMP-treated mice vs. -untreated mice. The functions of the common proteins modified by TMP in the two models were mainly involved in mitochondrial, synaptic, cytoskeleton, ATP binding, and GTP binding. Mitochondrial omics analysis revealed 21 and 20 differentially expressed mitochondrial proteins modified by TMP in 3xTg-AD mice and APP/PS1 mice, respectively. These differential proteins were located in the mitochondrial inner membrane, mitochondrial outer membrane, mitochondrial gap, and mitochondrial matrix, and the function of some proteins is closely related to oxidative phosphorylation (OXPHOS). Western-blot analysis confirmed that TMP changed the expression of OXPHOS complex proteins (sdhb, ndufa10, uqcrfs1, cox5b, atp5a) in the hippocampus of the two AD mice. Taken together, we demonstrated that TMP treatment changed the hippocampal proteome, reduced AD pathology, and reduced cognitive impairment in the two AD models. The changes might be associated with modification of the mitochondrial protein profile by TMP. The results of the study suggest that TMP can improve the symptoms of AD.
Collapse
Affiliation(s)
- Xianfeng Huang
- School of Pharmacy and School of Medicine, Changzhou University, Changzhou, China
| | - Jinyao Yang
- School of Pharmacy and School of Medicine, Changzhou University, Changzhou, China.,Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Subject of Modern Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Xi Huang
- Department of Neurology, Shenzhen People's Hospital (First Affiliated Hospital of Southern University of Science and Technology), Second Clinical College, Jinan University, Shenzhen, China
| | - Zaijun Zhang
- Institute of New Drug Research and Guangzhou, Key Laboratory of Innovative Chemical Drug Research in Cardio-Cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Jianjun Liu
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Subject of Modern Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Liangyu Zou
- Department of Neurology, Shenzhen People's Hospital (First Affiliated Hospital of Southern University of Science and Technology), Second Clinical College, Jinan University, Shenzhen, China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Subject of Modern Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| |
Collapse
|
7
|
Alghamdi F, Alshuweishi Y, Salt IP. Regulation of nutrient uptake by AMP-activated protein kinase. Cell Signal 2020; 76:109807. [DOI: 10.1016/j.cellsig.2020.109807] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 02/07/2023]
|
8
|
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
|
9
|
Rehman A, Hu SH, Tnimov Z, Whitten AE, King GJ, Jarrott RJ, Norwood SJ, Alexandrov K, Collins BM, Christie MP, Martin JL. The nature of the Syntaxin4 C-terminus affects Munc18c-supported SNARE assembly. PLoS One 2017; 12:e0183366. [PMID: 28841669 PMCID: PMC5571939 DOI: 10.1371/journal.pone.0183366] [Citation(s) in RCA: 4] [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: 03/06/2017] [Accepted: 08/02/2017] [Indexed: 11/18/2022] Open
Abstract
Vesicular transport of cellular cargo requires targeted membrane fusion and formation of a SNARE protein complex that draws the two apposing fusing membranes together. Insulin-regulated delivery and fusion of glucose transporter-4 storage vesicles at the cell surface is dependent on two key proteins: the SNARE integral membrane protein Syntaxin4 (Sx4) and the soluble regulatory protein Munc18c. Many reported in vitro studies of Munc18c:Sx4 interactions and of SNARE complex formation have used soluble Sx4 constructs lacking the native transmembrane domain. As a consequence, the importance of the Sx4 C-terminal anchor remains poorly understood. Here we show that soluble C-terminally truncated Sx4 dissociates more rapidly from Munc18c than Sx4 where the C-terminal transmembrane domain is replaced with a T4-lysozyme fusion. We also show that Munc18c appears to inhibit SNARE complex formation when soluble C-terminally truncated Sx4 is used but does not inhibit SNARE complex formation when Sx4 is C-terminally anchored (by a C-terminal His-tag bound to resin, by a C-terminal T4L fusion or by the native C-terminal transmembrane domain in detergent micelles). We conclude that the C-terminus of Sx4 is critical for its interaction with Munc18c, and that the reported inhibitory role of Munc18c may be an artifact of experimental design. These results support the notion that a primary role of Munc18c is to support SNARE complex formation and membrane fusion.
Collapse
Affiliation(s)
- Asma Rehman
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Shu-Hong Hu
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Zakir Tnimov
- Division of Cell Biology and Molecular Medicine, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Andrew E. Whitten
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Gordon J. King
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Russell J. Jarrott
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Suzanne J. Norwood
- Division of Cell Biology and Molecular Medicine, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Kirill Alexandrov
- Division of Cell Biology and Molecular Medicine, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Brett M. Collins
- Division of Cell Biology and Molecular Medicine, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Michelle P. Christie
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
- * E-mail: (MPC); (JLM)
| | - Jennifer L. Martin
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
- * E-mail: (MPC); (JLM)
| |
Collapse
|
10
|
Garcia NA, Moncayo-Arlandi J, Sepulveda P, Diez-Juan A. Cardiomyocyte exosomes regulate glycolytic flux in endothelium by direct transfer of GLUT transporters and glycolytic enzymes. Cardiovasc Res 2015; 109:397-408. [PMID: 26609058 DOI: 10.1093/cvr/cvv260] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 11/10/2015] [Indexed: 12/25/2022] Open
Abstract
AIMS Cardiomyocytes (CMs) and endothelial cells (ECs) have an intimate anatomical relationship, which is essential for maintaining the metabolic requirements of the heart. Little is known about the mechanisms that regulate nutrient flow from ECs to associated CMs, especially in situations of acute stress when local active processes are required to regulate endothelial transport. We examined whether CM-derived exosomes can modulate glucose transport and metabolism in ECs. METHODS AND RESULTS In conditions of glucose deprivation, CMs increase the synthesis and secretion of exosomes. These exosomes are loaded with functional glucose transporters and glycolytic enzymes, which are internalized by ECs, leading to increased glucose uptake, glycolytic activity, and pyruvate production in recipient cells. CONCLUSION These findings establish CM-derived exosomes as key components of the cardio-endothelial communication system which, through intercellular protein complementation, would allow a rapid response from ECs to increase glucose transport and a putative uptake of metabolic fuels from blood to CMs. This CM-EC protein complementation process might have implications for metabolic regulation in health and disease.
Collapse
Affiliation(s)
- Nahuel A Garcia
- Mixt Unit for Cardiovascular Repair, Instituto de Investigación Sanitaria La Fe-Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Javier Moncayo-Arlandi
- Cardiovascular Genetics Center, Institut d́Investigació Biomèdica de Girona, Girona, Spain
| | - Pilar Sepulveda
- Mixt Unit for Cardiovascular Repair, Instituto de Investigación Sanitaria La Fe-Centro de Investigación Príncipe Felipe, Valencia, Spain Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, Adva. Fernando Abril Martorell 106, 46026 Valencia, Spain
| | - Antonio Diez-Juan
- Fundación IVI/INCLIVA, Valencia, Spain IGENOMICS, Calle Catedrático Agustín Escardino 9, Paterna, Valencia 46980, Spain
| |
Collapse
|
11
|
Sadler JBA, Bryant NJ, Gould GW. Characterization of VAMP isoforms in 3T3-L1 adipocytes: implications for GLUT4 trafficking. Mol Biol Cell 2014; 26:530-6. [PMID: 25501368 PMCID: PMC4310743 DOI: 10.1091/mbc.e14-09-1368] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The levels of expression, distribution, and association of all of the VAMPs expressed in 3T3-L1 adipocytes are characterized. This is the first systematic analysis of all members of this protein family for any cell type. The fusion of GLUT4-containing vesicles with the plasma membrane of adipocytes is a key facet of insulin action. This process is mediated by the formation of functional soluble N-ethylmaleimide–sensitive factor attachment protein receptor (SNARE) complexes between the plasma membrane t-SNARE complex and the vesicle v-SNARE or VAMP. The t-SNARE complex consists of Syntaxin4 and SNAP23, and whereas many studies identify VAMP2 as the v-SNARE, others suggest that either VAMP3 or VAMP8 may also fulfil this role. Here we characterized the levels of expression, distribution, and association of all the VAMPs expressed in 3T3-L1 adipocytes to provide the first systematic analysis of all members of this protein family for any cell type. Despite our finding that all VAMP isoforms form SDS-resistant SNARE complexes with Syntaxin4/SNAP23 in vitro, a combination of levels of expression (which vary by >30-fold), subcellular distribution, and coimmunoprecipitation analyses lead us to propose that VAMP2 is the major v-SNARE involved in GLUT4 trafficking to the surface of 3T3-L1 adipocytes.
Collapse
Affiliation(s)
- Jessica B A Sadler
- Henry Wellcome Laboratory of Cell Biology, Institute for Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Nia J Bryant
- Department of Biology, University of York, Heslington YO10 5DD, United Kingdom
| | - Gwyn W Gould
- Henry Wellcome Laboratory of Cell Biology, Institute for Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| |
Collapse
|
12
|
Organization of organelles and VAMP-associated vesicular transport systems in differentiating skeletal muscle cells. Anat Sci Int 2014; 90:33-9. [DOI: 10.1007/s12565-014-0266-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 11/19/2014] [Indexed: 10/24/2022]
|
13
|
Caceres PS, Mendez M, Ortiz PA. Vesicle-associated membrane protein 2 (VAMP2) but Not VAMP3 mediates cAMP-stimulated trafficking of the renal Na+-K+-2Cl- co-transporter NKCC2 in thick ascending limbs. J Biol Chem 2014; 289:23951-62. [PMID: 25008321 PMCID: PMC4156046 DOI: 10.1074/jbc.m114.589333] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In the kidney, epithelial cells of the thick ascending limb (TAL) reabsorb NaCl via the apical Na+/K+/2Cl− co-transporter NKCC2. Steady-state surface NKCC2 levels in the apical membrane are maintained by a balance between exocytic delivery, endocytosis, and recycling. cAMP is the second messenger of hormones that enhance NaCl absorption. cAMP stimulates NKCC2 exocytic delivery via protein kinase A (PKA), increasing steady-state surface NKCC2. However, the molecular mechanism involved has not been studied. We found that several members of the SNARE family of membrane fusion proteins are expressed in TALs. Here we report that NKCC2 co-immunoprecipitates with VAMP2 in rat TALs, and they co-localize in discrete domains at the apical surface. cAMP stimulation enhanced VAMP2 exocytic delivery to the plasma membrane of renal cells, and stimulation of PKA enhanced VAMP2-NKCC2 co-immunoprecipitation in TALs. In vivo silencing of VAMP2 but not VAMP3 in TALs blunted cAMP-stimulated steady-state surface NKCC2 expression and completely blocked cAMP-stimulated NKCC2 exocytic delivery. VAMP2 was not involved in constitutive NKCC2 delivery. We concluded that VAMP2 but not VAMP3 selectively mediates cAMP-stimulated NKCC2 exocytic delivery and surface expression in TALs. We also demonstrated that cAMP stimulation enhances VAMP2 exocytosis and promotes VAMP2 interaction with NKCC2.
Collapse
Affiliation(s)
- Paulo S Caceres
- From the Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan 48202 and the Department of Physiology, Wayne State University, Detroit, Michigan 48202
| | - Mariela Mendez
- From the Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan 48202 and
| | - Pablo A Ortiz
- From the Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan 48202 and the Department of Physiology, Wayne State University, Detroit, Michigan 48202
| |
Collapse
|
14
|
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
|
15
|
Burchfield JG, Lu J, Fazakerley DJ, Tan SX, Ng Y, Mele K, Buckley MJ, Han W, Hughes WE, James DE. Novel systems for dynamically assessing insulin action in live cells reveals heterogeneity in the insulin response. Traffic 2013; 14:259-73. [PMID: 23252720 DOI: 10.1111/tra.12035] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 12/13/2012] [Accepted: 12/18/2012] [Indexed: 12/23/2022]
Abstract
Regulated GLUT4 trafficking is a key action of insulin. Quantitative stepwise analysis of this process provides a powerful tool for pinpointing regulatory nodes that contribute to insulin regulation and insulin resistance. We describe a novel GLUT4 construct and workflow for the streamlined dissection of GLUT4 trafficking; from simple high throughput screens to high resolution analyses of individual vesicles. We reveal single cell heterogeneity in insulin action highlighting the utility of this approach - each cell displayed a unique and highly reproducible insulin response, implying that each cell is hard-wired to produce a specific output in response to a given stimulus. These data highlight that the response of a cell population to insulin is underpinned by extensive heterogeneity at the single cell level. This heterogeneity is pre-programmed within each cell and is not the result of intracellular stochastic events.
Collapse
Affiliation(s)
- James G Burchfield
- Diabetes and Obesity Research Program, The Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
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
|
17
|
Abstract
Despite daily fasting and feeding, plasma glucose levels are normally maintained within a narrow range owing to the hormones insulin and glucagon. Insulin increases glucose uptake into fat and muscle cells through the regulated trafficking of vesicles that contain glucose transporter type 4 (GLUT4). New insights into insulin signalling reveal that phosphorylation events initiated by the insulin receptor regulate key GLUT4 trafficking proteins, including small GTPases, tethering complexes and the vesicle fusion machinery. These proteins, in turn, control GLUT4 movement through the endosomal system, formation and retention of specialized GLUT4 storage vesicles and targeted exocytosis of these vesicles. Understanding these processes may help to explain the development of insulin resistance in type 2 diabetes and provide new potential therapeutic targets.
Collapse
|
18
|
Reverter M, Rentero C, de Muga SV, Alvarez-Guaita A, Mulay V, Cairns R, Wood P, Monastyrskaya K, Pol A, Tebar F, Blasi J, Grewal T, Enrich C. Cholesterol transport from late endosomes to the Golgi regulates t-SNARE trafficking, assembly, and function. Mol Biol Cell 2012; 22:4108-23. [PMID: 22039070 PMCID: PMC3204072 DOI: 10.1091/mbc.e11-04-0332] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
This study shows that impaired cholesterol egress from late endosomes in cells with high annexin A6 levels is associated with altered soluble N-ethylmaleimide–sensitive fusion protein 23 (SNAP23) and syntaxin-4 cellular distribution and assembly and accumulation in Golgi membranes. This correlates with reduced secretion of cargo along the constitutive and SNAP23/syntaxin-4–dependent secretory pathway. Cholesterol regulates plasma membrane (PM) association and functioning of syntaxin-4 and soluble N-ethylmaleimide-sensitive fusion protein 23 (SNAP23) in the secretory pathway. However, the molecular mechanism and cellular cholesterol pools that determine the localization and assembly of these target membrane SNAP receptors (t-SNAREs) are largely unknown. We recently demonstrated that high levels of annexin A6 (AnxA6) induce accumulation of cholesterol in late endosomes, thereby reducing cholesterol in the Golgi and PM. This leads to an impaired supply of cholesterol needed for cytosolic phospholipase A2 (cPLA2) to drive Golgi vesiculation and caveolin transport to the cell surface. Using AnxA6-overexpressing cells as a model for cellular cholesterol imbalance, we identify impaired cholesterol egress from late endosomes and diminution of Golgi cholesterol as correlating with the sequestration of SNAP23/syntaxin-4 in Golgi membranes. Pharmacological accumulation of late endosomal cholesterol and cPLA2 inhibition induces a similar phenotype in control cells with low AnxA6 levels. Ectopic expression of Niemann-Pick C1 (NPC1) or exogenous cholesterol restores the location of SNAP23 and syntaxin-4 within the PM. Importantly, AnxA6-mediated mislocalization of these t-SNAREs correlates with reduced secretion of cargo via the SNAP23/syntaxin-4–dependent constitutive exocytic pathway. We thus conclude that inhibition of late endosomal export and Golgi cholesterol depletion modulate t-SNARE localization and functioning along the exocytic pathway.
Collapse
Affiliation(s)
- Meritxell Reverter
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Universitat de Barcelona, 08036 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Reverter M, Rentero C, de Muga SV, Alvarez-Guaita A, Mulay V, Cairns R, Wood P, Monastyrskaya K, Pol A, Tebar F, Blasi J, Grewal T, Enrich C. Cholesterol transport from late endosomes to the Golgi regulates t-SNARE trafficking, assembly, and function. Mol Biol Cell 2011. [DOI: 10.1091/mbc.e11-04-0332r] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Cholesterol regulates plasma membrane (PM) association and functioning of syntaxin-4 and soluble N-ethylmaleimide-sensitive fusion protein 23 (SNAP23) in the secretory pathway. However, the molecular mechanism and cellular cholesterol pools that determine the localization and assembly of these target membrane SNAP receptors (t-SNAREs) are largely unknown. We recently demonstrated that high levels of annexin A6 (AnxA6) induce accumulation of cholesterol in late endosomes, thereby reducing cholesterol in the Golgi and PM. This leads to an impaired supply of cholesterol needed for cytosolic phospholipase A2(cPLA2) to drive Golgi vesiculation and caveolin transport to the cell surface. Using AnxA6-overexpressing cells as a model for cellular cholesterol imbalance, we identify impaired cholesterol egress from late endosomes and diminution of Golgi cholesterol as correlating with the sequestration of SNAP23/syntaxin-4 in Golgi membranes. Pharmacological accumulation of late endosomal cholesterol and cPLA2inhibition induces a similar phenotype in control cells with low AnxA6 levels. Ectopic expression of Niemann-Pick C1 (NPC1) or exogenous cholesterol restores the location of SNAP23 and syntaxin-4 within the PM. Importantly, AnxA6-mediated mislocalization of these t-SNAREs correlates with reduced secretion of cargo via the SNAP23/syntaxin-4–dependent constitutive exocytic pathway. We thus conclude that inhibition of late endosomal export and Golgi cholesterol depletion modulate t-SNARE localization and functioning along the exocytic pathway.
Collapse
Affiliation(s)
- Meritxell Reverter
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Carles Rentero
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Sandra Vilà de Muga
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Anna Alvarez-Guaita
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Vishwaroop Mulay
- Faculty of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia
| | - Rose Cairns
- Faculty of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia
| | - Peta Wood
- Faculty of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia
| | - Katia Monastyrskaya
- Urology Research Laboratory, Department of Clinical Research, University of Bern, 3000 Bern 9, Switzerland
| | - Albert Pol
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Francesc Tebar
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Joan Blasi
- Department of Pathology and Experimental Therapeutics, IDIBELL–University of Barcelona, 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Thomas Grewal
- Faculty of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia
| | - Carlos Enrich
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
20
|
Lin Y, Sun Z. Thyroid hormone promotes insulin-induced glucose uptake by enhancing Akt phosphorylation and VAMP2 translocation in 3T3-L1 adipocytes. J Cell Physiol 2011; 226:2625-32. [PMID: 21792921 DOI: 10.1002/jcp.22613] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The purpose of this study was to test a hypothesis that T3 promotes glucose uptake via enhancing insulin-induced Akt phosphorylation and VAMP2 translocation in 3T3-L1 adipocytes. T3 significantly enhanced insulin-induced phosphorylation of Akt, cytoplasma to cell membrane translocations of vesicle-associated membrane protein 2 (VAMP2) and glucose transporter 4 (GLUT4), and glucose uptake in adipocytes. Akt inhibitor X abolished the promoting effects of T3, suggesting that Akt activation is essential for T3 to enhance these insulin-induced events in adipocytes. Knockdown of VAMP2 using siRNA abrogated the effects of T3 on insulin-induced GLUT4 translocation and glucose uptake, suggesting that VAMP2 is an important mediator of these processes. These data suggest that T3 may promote glucose uptake via enhancing insulin-induced phosphorylation of Akt and subsequent translocations of VAMP2 and GLUT4 in 3T3-L1 adipocytes. Akt phosphorylation is necessary for the promoting effects of T3 on insulin-stimulated VAMP2 translocation. Further, VAMP2 is essential for T3 to increase insulin-stimulated translocation of GLUT4 and subsequent uptake of glucose in adipocytes.
Collapse
Affiliation(s)
- Yi Lin
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73126-0901, USA
| | | |
Collapse
|
21
|
Mendez M, Gross KW, Glenn ST, Garvin JL, Carretero OA. Vesicle-associated membrane protein-2 (VAMP2) mediates cAMP-stimulated renin release in mouse juxtaglomerular cells. J Biol Chem 2011; 286:28608-18. [PMID: 21708949 PMCID: PMC3151102 DOI: 10.1074/jbc.m111.225839] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Renin is essential for blood pressure control. Renin is stored in granules in juxtaglomerular (JG) cells, located in the pole of the renal afferent arterioles. The second messenger cAMP stimulates renin release. However, it is unclear whether fusion and exocytosis of renin-containing granules is involved. In addition, the role of the fusion proteins, SNAREs (soluble N-ethylmaleimide-sensitive factor attachment proteins), in renin release from JG cells has not been studied. The vesicle SNARE proteins VAMP2 (vesicle associated membrane protein 2) and VAMP3 mediate cAMP-stimulated exocytosis in other endocrine cells. Thus, we hypothesized that VAMP2 and/or -3 mediate cAMP-stimulated renin release from JG cells. By fluorescence-activated cell sorting, we isolated JG cells expressing green fluorescent protein and compared the relative abundance of VAMP2/3 in JG cells versus total mouse kidney mRNA by quantitative PCR. We found that VAMP2 and VAMP3 mRNA are expressed and enriched in JG cells. Confocal imaging of primary cultures of JG cells showed that VAMP2 (but not VAMP3) co-localized with renin-containing granules. Cleavage of VAMP2 and VAMP3 with tetanus toxin blocked cAMP-stimulated renin release from JG cells by ∼50% and impaired cAMP-stimulated exocytosis by ∼50%, as monitored with FM1–43. Then we specifically knocked down VAMP2 or VAMP3 by adenoviral-mediated delivery of short hairpin silencing RNA. We found that silencing VAMP2 blocked cAMP-induced renin release by ∼50%. In contrast, silencing VAMP3 had no effect on basal or cAMP-stimulated renin release. We conclude that VAMP2 and VAMP3 are expressed in JG cells, but only VAMP2 is targeted to renin-containing granules and mediates the stimulatory effect of cAMP on renin exocytosis.
Collapse
Affiliation(s)
- Mariela Mendez
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan 48202, USA.
| | | | | | | | | |
Collapse
|
22
|
Abstract
Delivery of the glucose transporter type 4 (GLUT4) from an intracellular location to the cell surface in response to insulin represents a specialized form of membrane traffic, known to be impaired in the disease states of insulin resistance and type 2 diabetes. Like all membrane trafficking events, this translocation of GLUT4 requires members of the SNARE family of proteins. Here, we discuss two SNARE complexes that have been implicated in insulin-regulated GLUT4 traffic: one regulating the final delivery of GLUT4 to the cell surface in response to insulin and the other controlling GLUT4's intracellular trafficking.
Collapse
Affiliation(s)
- Nia J Bryant
- Henry Wellcome Laboratory of Cell Biology, Institute of Molecular, Cell and Systems Biology, Davidson Building, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | | |
Collapse
|
23
|
Subcellular trafficking of the substrate transporters GLUT4 and CD36 in cardiomyocytes. Cell Mol Life Sci 2011; 68:2525-38. [PMID: 21547502 PMCID: PMC3134709 DOI: 10.1007/s00018-011-0690-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2011] [Revised: 04/01/2011] [Accepted: 04/12/2011] [Indexed: 12/16/2022]
Abstract
Cardiomyocytes use glucose as well as fatty acids for ATP production. These substrates are transported into the cell by glucose transporter 4 (GLUT4) and the fatty acid transporter CD36. Besides being located at the sarcolemma, GLUT4 and CD36 are stored in intracellular compartments. Raised plasma insulin concentrations and increased cardiac work will stimulate GLUT4 as well as CD36 to translocate to the sarcolemma. As so far studied, signaling pathways that regulate GLUT4 translocation similarly affect CD36 translocation. During the development of insulin resistance and type 2 diabetes, CD36 becomes permanently localized at the sarcolemma, whereas GLUT4 internalizes. This juxtaposed positioning of GLUT4 and CD36 is important for aberrant substrate uptake in the diabetic heart: chronically increased fatty acid uptake at the expense of glucose. To explain the differences in subcellular localization of GLUT4 and CD36 in type 2 diabetes, recent research has focused on the role of proteins involved in trafficking of cargo between subcellular compartments. Several of these proteins appear to be similarly involved in both GLUT4 and CD36 translocation. Others, however, have different roles in either GLUT4 or CD36 translocation. These trafficking components, which are differently involved in GLUT4 or CD36 translocation, may be considered novel targets for the development of therapies to restore the imbalanced substrate utilization that occurs in obesity, insulin resistance and diabetic cardiomyopathy.
Collapse
|
24
|
Hasan N, Corbin D, Hu C. Fusogenic pairings of vesicle-associated membrane proteins (VAMPs) and plasma membrane t-SNAREs--VAMP5 as the exception. PLoS One 2010; 5:e14238. [PMID: 21151919 PMCID: PMC2997805 DOI: 10.1371/journal.pone.0014238] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Accepted: 11/12/2010] [Indexed: 12/31/2022] Open
Abstract
Background Intracellular vesicle fusion is mediated by the interactions of SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) proteins on vesicles (v-SNAREs) and on target membranes (t-SNAREs). The vesicle-associated membrane proteins (VAMPs) are v-SNAREs that reside in various post-Golgi vesicular compartments. To fully understand the specific role of each VAMP in vesicle trafficking, it is important to determine if VAMPs have differential membrane fusion activities. Methodology/Principal Findings In this study, we developed a cell fusion assay that quantifies SNARE-mediated membrane fusion events by activated expression of β-galactosidase, and examined fusogenic pairings between the seven VAMPs, i.e., VAMPs 1, 2, 3, 4, 5, 7 and 8, and two plasma membrane t-SNARE complexes, syntaxin1/SNAP-25 and syntaxin4/SNAP-25. VAMPs 1, 2, 3, 4, 7 and 8 drove fusion efficiently, whereas VAMP5 was unable to mediate fusion with the t-SNAREs. By expressing VAMPs 1, 3, 4, 7 and 8 at the same level, we further compared their membrane fusion activities. VAMPs 1 and 3 had comparable and the highest fusion activities, whereas VAMPs 4, 7 and 8 exhibited 30–50% lower fusion activities. Moreover, we determined the dependence of cell fusion activity on VAMP1 expression level. Analysis of the dependence data suggested that there was no cooperativity of VAMP proteins in the cell fusion reaction. Conclusions/Significance These data indicate that VAMPs have differential membrane fusion capacities, and imply that with the exception of VAMP5, VAMPs are essentially redundant in mediating fusion with plasma membrane t-SNAREs.
Collapse
Affiliation(s)
- Nazarul Hasan
- Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Deborah Corbin
- Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Chuan Hu
- Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
25
|
Stimulation of GLUT4 (glucose transporter isoform 4) storage vesicle formation by sphingolipid depletion. Biochem J 2010; 427:143-50. [PMID: 20085539 DOI: 10.1042/bj20091529] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Insulin stimulates glucose transport in fat and skeletal muscle cells primarily by inducing the translocation of GLUT4 (glucose transporter isoform 4) to the PM (plasma membrane) from specialized GSVs (GLUT4 storage vesicles). Glycosphingolipids are components of membrane microdomains and are involved in insulin-regulated glucose transport. Cellular glycosphingolipids decrease during adipocyte differentiation and have been suggested to be involved in adipocyte function. In the present study, we investigated the role of glycosphingolipids in regulating GLUT4 translocation. We decreased glycosphingolipids in 3T3-L1 adipocytes using glycosphingolipid synthesis inhibitors and investigated the effects on GLUT4 translocation using immunocytochemistry, preparation of PM sheets, isolation of GSVs and FRAP (fluorescence recovery after photobleaching) of GLUT4-GFP (green fluorescent protein) in intracellular structures. Glycosphingolipids were located in endosomal vesicles in pre-adipocytes and redistributed to the PM with decreased expression at day 2 after initiation of differentiation. In fully differentiated adipocytes, depletion of glycosphingolipids dramatically accelerated insulin-stimulated GLUT4 translocation. Although insulin-induced phosphorylation of IRS (insulin receptor substrate) and Akt remained intact in glycosphingolipid-depleted cells, both in vitro budding of GLUT4 vesicles and FRAP of GLUT4-GFP on GSVs were stimulated. Glycosphingolipid depletion also enhanced the insulin-induced translocation of VAMP2 (vesicle-associated membrane protein 2), but not the transferrin receptor or cellubrevin, indicating that the effect of glycosphingolipids was specific to VAMP2-positive GSVs. Our results strongly suggest that decreasing glycosphingolipid levels promotes the formation of GSVs and, thus, GLUT4 translocation. These studies provide a mechanistic basis for recent studies showing that inhibition of glycosphingolipid synthesis improves glycaemic control and enhances insulin sensitivity in animal models of Type 2 diabetes.
Collapse
|
26
|
Jewell JL, Oh E, Thurmond DC. Exocytosis mechanisms underlying insulin release and glucose uptake: conserved roles for Munc18c and syntaxin 4. Am J Physiol Regul Integr Comp Physiol 2010; 298:R517-31. [PMID: 20053958 DOI: 10.1152/ajpregu.00597.2009] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Type 2 diabetes has been coined "a two-hit disease," as it involves specific defects of glucose-stimulated insulin secretion from the pancreatic beta cells in addition to defects in peripheral tissue insulin action required for glucose uptake. Both of these processes, insulin secretion and glucose uptake, are mediated by SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) protein core complexes composed of syntaxin, SNAP-23/25, and VAMP proteins. The SNARE core complex is regulated by the Sec1/Munc18 (SM) family of proteins, which selectively bind to their cognate syntaxin isoforms with high affinity. The process of insulin secretion uses multiple Munc18-syntaxin isoform pairs, whereas insulin action in the peripheral tissues appears to use only the Munc18c-syntaxin 4 pair. Importantly, recent reports have linked obesity and Type 2 diabetes in humans with changes in protein levels and single nucleotide polymorphisms (SNPs) of Munc18 and syntaxin isoforms relevant to these exocytotic processes, although the molecular mechanisms underlying the observed phenotypes remain incomplete (5, 104, 144). Given the conservation of these proteins in two seemingly disparate processes and the need to design and implement novel and more effective clinical interventions, it will be vitally important to delineate the mechanisms governing these conserved SNARE-mediated exocytosis events. Thus, we provide here an up-to-date historical review of advancements in defining the roles and molecular mechanisms of Munc18-syntaxin complexes in the pathophysiology of Type 2 diabetes.
Collapse
Affiliation(s)
- Jenna L Jewell
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | |
Collapse
|
27
|
Hasan N, Hu C. Vesicle-associated membrane protein 2 mediates trafficking of α5β1 integrin to the plasma membrane. Exp Cell Res 2010; 316:12-23. [DOI: 10.1016/j.yexcr.2009.10.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Revised: 09/02/2009] [Accepted: 10/05/2009] [Indexed: 01/11/2023]
|
28
|
Kawaguchi T, Tamori Y, Kanda H, Yoshikawa M, Tateya S, Nishino N, Kasuga M. The t-SNAREs syntaxin4 and SNAP23 but not v-SNARE VAMP2 are indispensable to tether GLUT4 vesicles at the plasma membrane in adipocyte. Biochem Biophys Res Commun 2010; 391:1336-41. [DOI: 10.1016/j.bbrc.2009.12.045] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Accepted: 12/10/2009] [Indexed: 12/31/2022]
|
29
|
Zhao P, Yang L, Lopez JA, Fan J, Burchfield JG, Bai L, Hong W, Xu T, James DE. Variations in the requirement for v-SNAREs in GLUT4 trafficking in adipocytes. J Cell Sci 2009; 122:3472-80. [PMID: 19759285 DOI: 10.1242/jcs.047449] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Vesicle transport in eukaryotic cells is regulated by SNARE proteins, which play an intimate role in regulating the specificity of vesicle fusion between discrete intracellular organelles. In the present study we investigated the function and plasticity of v-SNAREs in insulin-regulated GLUT4 trafficking in adipocytes. Using a combination of knockout mice, v-SNARE cleavage by clostridial toxins and total internal reflection fluorescence microscopy, we interrogated the function of VAMPs 2, 3 and 8 in this process. Our studies reveal that the simultaneous disruption of VAMPs 2, 3 and 8 completely inhibited insulin-stimulated GLUT4 insertion into the plasma membrane, due to a block in vesicle docking at the plasma membrane. These defects could be rescued by re-expression of VAMP2, VAMP3 or VAMP8 alone, but not VAMP7. These data indicate a plasticity in the requirement for v-SNAREs in GLUT4 trafficking to the plasma membrane and further define an important role for the v-SNARE proteins in pre-fusion docking of vesicles.
Collapse
Affiliation(s)
- Ping Zhao
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Down-regulating protein kinase C alpha: functional cooperation between the proteasome and the endocytic system. Cell Signal 2009; 21:1607-19. [PMID: 19586612 DOI: 10.1016/j.cellsig.2009.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Revised: 05/31/2009] [Accepted: 06/26/2009] [Indexed: 11/23/2022]
Abstract
Ubiquitination, proteasome, caveolae and endosomes have been implicated in controlling protein kinase C alpha (PKC alpha) down-regulation. However, the molecular mechanism remained obscure. Here we show that endosomes and proteasome cooperate in phorbol ester 12-O-tetradecanoyl phorbol acetate (TPA)-induced down-regulation of PKC alpha. We show that following TPA treatment and translocation to the plasma membrane, PKC alpha undergoes multimonoubiquitination prior to its degradation by the proteasome. However, to reach the proteasome, PKC alpha must travel through the endocytic system from early to late endosomes. This route requires functional endosomes, whereby endosomal alkalinization, or ablation, abrogates completely PKC alpha degradation maintaining the enzyme at the plasma membrane. This route also depends on synaptotagmin (Syt) II and the Rab7 GTPase, whereby Syt II knock-down or expression of the GDP-locked Rab7 inactive mutant prevents PKC alpha degradation. We further show that proteasome plays a dual role, where an active proteasome is required for deubiquitination of PKC alpha, a step crucial to prevent PKC alpha targeting to the endocytic recycling compartment. Finally, we show that the association with rafts-localized cell surface proteins that internalize in a clathrin-independent fashion is necessary to allow the trafficking of PKC alpha from the plasma membrane to the proteasome, its ultimate degradation station.
Collapse
|
31
|
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: 2.2] [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
|
32
|
Yip MF, Ramm G, Larance M, Hoehn KL, Wagner MC, Guilhaus M, James DE. CaMKII-mediated phosphorylation of the myosin motor Myo1c is required for insulin-stimulated GLUT4 translocation in adipocytes. Cell Metab 2008; 8:384-98. [PMID: 19046570 DOI: 10.1016/j.cmet.2008.09.011] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2007] [Revised: 03/02/2008] [Accepted: 09/19/2008] [Indexed: 11/26/2022]
Abstract
The unconventional myosin Myo1c has been implicated in insulin-regulated GLUT4 translocation to the plasma membrane in adipocytes. We show that Myo1c undergoes insulin-dependent phosphorylation at S701. Phosphorylation was accompanied by enhanced 14-3-3 binding and reduced calmodulin binding. Recombinant CaMKII phosphorylated Myo1c in vitro and siRNA knockdown of CaMKIIdelta abolished insulin-dependent Myo1c phosphorylation in vivo. CaMKII activity was increased upon insulin treatment and the CaMKII inhibitors CN21 and KN-62 or the Ca(2+) chelator BAPTA-AM blocked insulin-dependent Myo1c phosphorylation and insulin-stimulated glucose transport in adipocytes. Myo1c ATPase activity was increased after CaMKII phosphorylation in vitro and after insulin stimulation of CHO/IR/IRS-1 cells. Expression of wild-type Myo1c, but not S701A or ATPase dead mutant K111A, rescued the inhibition of GLUT4 translocation by siRNA-mediated Myo1c knockdown. These data suggest that insulin regulates Myo1c function via CaMKII-dependent phosphorylation, and these events play a role in insulin-regulated GLUT4 trafficking in adipocytes likely involving Myo1c motor activity.
Collapse
Affiliation(s)
- Ming Fai Yip
- Diabetes and Obesity Research Program, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | | | | | | | | | | | | |
Collapse
|
33
|
Pooley RD, Moynihan KL, Soukoulis V, Reddy S, Francis R, Lo C, Ma LJ, Bader DM. Murine CENPF interacts with syntaxin 4 in the regulation of vesicular transport. J Cell Sci 2008; 121:3413-21. [PMID: 18827011 DOI: 10.1242/jcs.032847] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Syntaxin 4 is a component of the SNARE complex that regulates membrane docking and fusion. Using a yeast two-hybrid screen, we identify a novel interaction between syntaxin 4 and cytoplasmic murine CENPF, a protein previously demonstrated to associate with the microtubule network and SNAP-25. The binding domain for syntaxin 4 in CENPF was defined by yeast two-hybrid assay and co-immunoprecipitation. Confocal analyses in cell culture reveal a high degree of colocalization between endogenously expressed proteins in interphase cells. Additionally, the endogenous SNARE proteins can be isolated as a complex with CENPF in immunoprecipitation experiments. Further analyses demonstrate that murine CENPF and syntaxin 4 colocalize with components of plasma membrane recycling: SNAP-25 and VAMP2. Depletion of endogenous CENPF disrupts GLUT4 trafficking whereas expression of a dominant-negative form of CENPF inhibits cell coupling. Taken together, these studies demonstrate that CENPF provides a direct link between proteins of the SNARE system and the microtubule network and indicate a diverse role for murine CENPF in vesicular transport.
Collapse
Affiliation(s)
- Ryan D Pooley
- Stahlman Cardiovascular Research Laboratories, Program for Developmental Biology, and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6300, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Williams D, Pessin JE. Mapping of R-SNARE function at distinct intracellular GLUT4 trafficking steps in adipocytes. ACTA ACUST UNITED AC 2008; 180:375-87. [PMID: 18227281 PMCID: PMC2213575 DOI: 10.1083/jcb.200709108] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The functional trafficking steps used by soluble NSF attachment protein receptor (SNARE) proteins have been difficult to establish because of substantial overlap in subcellular localization and because in vitro SNARE-dependent binding and fusion reactions can be promiscuous. Therefore, to functionally identify the site of action of the vesicle-associated membrane protein (VAMP) family of R-SNAREs, we have taken advantage of the temporal requirements of adipocyte biosynthetic sorting of a dual-tagged GLUT4 reporter (myc-GLUT4-GFP) coupled with small interfering RNA gene silencing. Using this approach, we confirm the requirement of VAMP2 and VAMP7 for insulin and osmotic shock trafficking from the vesicle storage sites, respectively, and fusion with the plasma membrane. Moreover, we identify a requirement for VAMP4 for the initial biosynthetic entry of GLUT4 from the Golgi apparatus into the insulin-responsive vesicle compartment, VAMP8, for plasma membrane endocytosis and VAMP2 for sorting to the specialized insulin-responsive compartment after plasma membrane endocytosis.
Collapse
Affiliation(s)
- Dumaine Williams
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| | | |
Collapse
|
35
|
Thurmond DC. Regulation of Insulin Action and Insulin Secretion by SNARE-Mediated Vesicle Exocytosis. MECHANISMS OF INSULIN ACTION 2007:52-70. [DOI: 10.1007/978-0-387-72204-7_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
36
|
Okayama M, Arakawa T, Mizoguchi I, Tajima Y, Takuma T. SNAP-23 is not essential for constitutive exocytosis in HeLa cells. FEBS Lett 2007; 581:4583-8. [PMID: 17825825 DOI: 10.1016/j.febslet.2007.08.046] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2007] [Revised: 08/09/2007] [Accepted: 08/21/2007] [Indexed: 01/28/2023]
Abstract
We applied the small interfering RNA (siRNA) technique and over-expression of a dominant-negative mutant to evaluate the role of SNAP-23, a non-neuronal isoform of SNAP-25, in constitutive exocytosis from HeLa cells. Although the protein level of SNAP-23 was reduced to less than 10% of the control value by siRNA directed against SNAP-23, exocytosis of SEAP (secreted alkaline phosphatase) was normal. Double knockdown of SNAP-23 and syntaxin-4 also failed to inhibit the secretion. Furthermore, over-expression of deltaC8-SNAP-23, a dominant-negative mutant of SNAP-23, did not abrogate SEAP secretion. These results suggest that SNAP-23 is not essential for constitutive exocytosis of SEAP.
Collapse
Affiliation(s)
- Miki Okayama
- Department of Orthodontics, School of Dentistry, Health Sciences University of Hokkaido, Tobetsu, Hokkaido 061-0293, Japan
| | | | | | | | | |
Collapse
|
37
|
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: 3.3] [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
|
38
|
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
|
39
|
Hatanaka T, Hatanaka Y, Tsuchida JI, Ganapathy V, Setou M. Amino acid transporter ATA2 is stored at the trans-Golgi network and released by insulin stimulus in adipocytes. J Biol Chem 2006; 281:39273-84. [PMID: 17050538 DOI: 10.1074/jbc.m604534200] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Recently, we cloned the ATA/SNAT transporters responsible for amino acid transport system A. System A is one of the major transport systems for small neutral and glucogenic amino acids represented by alanine and is involved in the metabolism of glucose and fat. Here, we describe the cellular mechanisms that participate in the acute translocation of ATA2 by insulin stimulus in 3T3-L1 adipocytes. We monitored this insulin-stimulated translocation of ATA2 using an expression system of enhanced green fluorescent protein-tagged ATA2. Studies in living cells revealed that ATA2 is stored in a discrete perinuclear site and that the transporter is released in vesicles from this site toward the plasma membrane. In immunofluorescent analysis, the storage site of ATA2 overlapped with the location of syntaxin 6, a marker of the trans-Golgi network (TGN), but not with that of EEA1, a marker of the early endosomes. The ATA2-containing vesicles on or near the plasma membrane were distinct from GLUT4-containing vesicles. Brefeldin A, an inhibitor of vesicular exit from the TGN, caused morphological changes in the ATA2 storage site along with the similar changes in the TGN. In non-transfected adipocytes, brefeldin A inhibited insulin-stimulated uptake of alpha-(methylamino)isobutyric acid more profoundly than insulin-stimulated uptake of 2-deoxy-d-glucose. These data demonstrate that the ATA2 storage site is specifically associated with the TGN and not with the general endosomal recycling system. Thus, the insulin-stimulated translocation pathways for ATA2 and GLUT4 in adipocytes are distinct, involving different storage sites.
Collapse
Affiliation(s)
- Takahiro Hatanaka
- Mitsubishi Kagaku Institute of Life Sciences (MITILS), 11 Minamiooya, Machida, Tokyo 194-8511, Japan
| | | | | | | | | |
Collapse
|
40
|
Proctor KM, Miller SCM, Bryant NJ, Gould GW. Syntaxin 16 controls the intracellular sequestration of GLUT4 in 3T3-L1 adipocytes. Biochem Biophys Res Commun 2006; 347:433-8. [PMID: 16828707 DOI: 10.1016/j.bbrc.2006.06.135] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2006] [Accepted: 06/16/2006] [Indexed: 11/22/2022]
Abstract
The regulated delivery of Glut4-containing vesicles to the plasma membrane is a specialised example of regulated membrane trafficking. Present models favour the transporter trafficking through two inter-related endosomal cycles. The first is the proto-typical endosomal system. This is a fast trafficking event that, in the absence of insulin, serves to internalise Glut4 from the plasma membrane. Once in this pathway, Glut4 is further sorted into a slowly recycling pathway that operates between recycling endosomes, the trans Golgi network, and a population of vesicles often referred to as Glut4-storage vesicles. Little is known about the molecules that regulate these distinct sorting steps. Here, we have studied the role of Stx16 in Glut4 trafficking. Using two independent strategies, we show that Stx16 plays a crucial role in Glut4 traffic in 3T3-L1 adipocytes. Over-expression of a mutant form of Stx16 devoid of a transmembrane anchor was found to significantly slow the reversal of insulin-stimulated glucose transport. Depletion of Stx16 using antisense approaches profoundly reduced insulin-stimulated glucose transport but was without effect on cell surface transferrin receptor levels, and also reduced the extent of Glut4 translocation to the plasma membrane in response to insulin. These data support a model in which Stx16 is crucial in the sorting of Glut4 from the fast cycling to the slow cycling intracellular trafficking pathways in adipocytes.
Collapse
Affiliation(s)
- Kirsty M Proctor
- Henry Wellcome Laboratory of Cell Biology, Division of Biochemistry and Molecular Biology, University of Glasgow, Davidson Building, Glasgow G12 8QQ, UK
| | | | | | | |
Collapse
|
41
|
Choudhury A, Marks DL, Proctor KM, Gould GW, Pagano RE. Regulation of caveolar endocytosis by syntaxin 6-dependent delivery of membrane components to the cell surface. Nat Cell Biol 2006; 8:317-28. [PMID: 16565709 DOI: 10.1038/ncb1380] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2005] [Accepted: 02/13/2006] [Indexed: 12/18/2022]
Abstract
Caveolar endocytosis has an important function in the cellular uptake of some bacterial toxins, viruses and circulating proteins. However, the molecular machinery involved in regulating caveolar uptake is poorly defined. Here, we demonstrate that caveolar endocytosis is regulated by syntaxin 6, a target membrane soluble N-ethylmaleimide attachment protein receptor (t-SNARE) involved in membrane fusion events along the secretory pathway. When syntaxin 6 function was inhibited, internalization through caveolae was dramatically reduced, whereas other endocytic mechanisms were unaffected. Syntaxin 6 inhibition also reduced the presence of caveolin-1 and caveolae at the plasma membrane. In addition, syntaxin 6 inhibition decreased the delivery of GM1 ganglioside (GM1) and glycosylphosphatidylinositol (GPI)-GFP (but not vesicular stomatitis virus-glycoprotein G; VSV-G) protein from the Golgi complex to the plasma membrane. Addition of GM1 to syntaxin 6-inhibited cells resulted in the reappearance of caveolin-1 and caveolae at the plasma membrane, and restored caveolar uptake. These results suggest that syntaxin 6 regulates the delivery of microdomain-associated lipids and proteins to the cell surface, which are required for caveolar endocytosis.
Collapse
Affiliation(s)
- Amit Choudhury
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, 200 First Street, S.W., Rochester, MN 55905, USA
| | | | | | | | | |
Collapse
|
42
|
Karpova AY, Tervo DGR, Gray NW, Svoboda K. Rapid and reversible chemical inactivation of synaptic transmission in genetically targeted neurons. Neuron 2006; 48:727-35. [PMID: 16337911 DOI: 10.1016/j.neuron.2005.11.015] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2004] [Revised: 05/05/2005] [Accepted: 11/08/2005] [Indexed: 10/25/2022]
Abstract
Inducible and reversible silencing of selected neurons in vivo is critical to understanding the structure and dynamics of brain circuits. We have developed Molecules for Inactivation of Synaptic Transmission (MISTs) that can be genetically targeted to allow the reversible inactivation of neurotransmitter release. MISTs consist of modified presynaptic proteins that interfere with the synaptic vesicle cycle when crosslinked by small molecule "dimerizers." MISTs based on the vesicle proteins VAMP2/Synaptobrevin and Synaptophysin induced rapid ( approximately 10 min) and reversible block of synaptic transmission in cultured neurons and brain slices. In transgenic mice expressing MISTs selectively in Purkinje neurons, administration of dimerizer reduced learning and performance of the rotarod behavior. MISTs allow for specific, inducible, and reversible lesions in neuronal circuits and may provide treatment of disorders associated with neuronal hyperactivity.
Collapse
|
43
|
Liao H, Keller SR, Castle JD. Insulin-Regulated Aminopeptidase Marks an Antigen-Stimulated Recycling Compartment in Mast Cells. Traffic 2006; 7:155-67. [PMID: 16420524 DOI: 10.1111/j.1600-0854.2006.00373.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Insulin-regulated aminopeptidase (IRAP) is a marker for insulin-sensitive recycling compartments of fat and muscle cells that contain the glucose transporter isoform GLUT4. Unlike GLUT4, IRAP is expressed in many other cell types. Thus, it is a potential marker for regulated recycling compartments that are analogous to GLUT4 vesicles. In bone marrow-derived mast cells, IRAP is highly expressed and localizes to an intracellular compartment different from secretory granules. Using cell-surface biotinylation, we determined that IRAP underwent rapid redistribution to the plasma membrane on antigen/immunoglobulin E (IgE) stimulation and was re-internalized within 30 min. When granule exocytosis was inhibited, by removing extracellular calcium, adding the protein kinase C inhibitor bisindolylmaleimide or the phosphatidylinositol 3-kinase inhibitor wortmannin, IRAP redistribution was still detected in stimulated cells. However, the redistribution of IRAP required intracellular calcium. By immunofluorescence, IRAP significantly co-localized with the transferrin receptor (TfR), a marker for constitutively recycling endosomes. However, antigen/IgE stimulation did not increase TfR on the cell surface, indicating that IRAP and TfR may follow different pathways to the plasma membrane. In rat peritoneal mast cells, the distributions of IRAP and TfR overlapped to only a limited extent, indicating that overlap may decrease with cell differentiation. We propose that IRAP vesicles represent a second IgE-sensitive exocytotic compartment in mast cells, which is regulated differently from secretory granules, and that these vesicles may be similar to GLUT4 vesicles.
Collapse
Affiliation(s)
- Haini Liao
- Department of Cell Biology, University of Virginia Health System, School of Medicine, Charlottesville, VA 22908, USA.
| | | | | |
Collapse
|
44
|
Dugani CB, Klip A. Glucose transporter 4: cycling, compartments and controversies. EMBO Rep 2005; 6:1137-42. [PMID: 16319959 PMCID: PMC1369215 DOI: 10.1038/sj.embor.7400584] [Citation(s) in RCA: 185] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2005] [Accepted: 10/19/2005] [Indexed: 11/08/2022] Open
Abstract
Insulin promotes glucose uptake into muscle and adipose tissues through glucose transporter 4 (GLUT4). In unstimulated cells, rapid endocytosis, slow exocytosis and dynamic or static retention cause GLUT4 to concentrate in early recycling endosomes, the trans-Golgi network and vesicle-associated protein 2-containing vesicles. The coordinated action of phosphatidylinositol 3-kinase effectors, protein kinase Akt, atypical protein kinase C (aPKC) and Akt substrate of 160-kDa (AS160), regulates the GLUT4 cycle by affecting its translocation, fusion with the plasma membrane, internalization and sorting. We review the evidence that supports such cycling, evaluate current models proposing static or dynamic retention, and highlight how distinct steps of GLUT4 transport are regulated by insulin signals. In particular, fusion seems to be regulated by aPKC (via munc18) and Akt (via syntaxin4-interacting protein (synip)). AS160 participates in GLUT4 intracellular retention, and possibly fusion, through candidate ras-related GTP-binding protein (Rab)2, Rab8, Rab10 and/or Rab14. The localization of the insulin-sensitive GLUT4 compartment and the precise target of insulin-derived signals remain open for future investigation.
Collapse
Affiliation(s)
- Chandrasagar B Dugani
- Programme in Cell Biology, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario M5G 1X8, Canada
| | - Amira Klip
- Department of Biochemistry, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
45
|
Liu G, Hou JC, Watson RT, Pessin JE. Initial entry of IRAP into the insulin-responsive storage compartment occurs prior to basal or insulin-stimulated plasma membrane recycling. Am J Physiol Endocrinol Metab 2005; 289:E746-52. [PMID: 15928022 DOI: 10.1152/ajpendo.00175.2005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To examine the acquisition of insulin sensitivity after the initial biosynthesis of the insulin-responsive aminopeptidase (IRAP), 3T3-L1 adipocytes were transfected with an enhanced green fluorescent protein-IRAP (EGFP-IRAP) fusion protein. In the absence of insulin, IRAP was rapidly localized (1-3 h) to secretory membranes and retained in these intracellular membrane compartments with little accumulation at the plasma membrane. However, insulin was unable to induce translocation to the plasma membrane until 6-9 h after biosynthesis. This was in marked contrast to another type II membrane protein (syntaxin 3) that rapidly defaulted to the plasma membrane 3 h after expression. In parallel with the time-dependent acquisition of insulin responsiveness, the newly synthesized IRAP protein converted from a brefeldin A-sensitive to a brefeldin A-insensitive state. The initial trafficking of IRAP to the insulin-responsive compartment was independent of plasma membrane endocytosis, as expression of a dominant-interfering dynamin mutant (Dyn/K44A) inhibited transferrin receptor endocytosis but had no effect on the insulin-stimulated translocation of the newly synthesized IRAP protein.
Collapse
Affiliation(s)
- Gang Liu
- Dept. of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | | | | | | |
Collapse
|
46
|
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: 8.1] [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
|
47
|
Uhlig M, Passlack W, Eckel J. Functional role of Rab11 in GLUT4 trafficking in cardiomyocytes. Mol Cell Endocrinol 2005; 235:1-9. [PMID: 15866422 DOI: 10.1016/j.mce.2005.02.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2004] [Revised: 02/17/2005] [Accepted: 02/26/2005] [Indexed: 01/15/2023]
Abstract
We have recently shown the co-localization of Rab11 and the glucose transporter GLUT4 in cardiac muscle and an insulin-stimulated increase of Rab11 in GLUT4-containing vesicles in this tissue. We now assessed the effect of Rab11 wt and a dominant-negative mutant (N124I) on GLUT4 trafficking in the cardiomyoblast cell line H9c2 stably overexpressing the insulin receptor (H9c2-E2) and in human primary skeletal myotubes. These cells were used for transient cotransfection or adenoviral co-infection with GLUT4myc and Rab11 wt or N124I with subsequent determination of 2-deoxyglucose (2-DOG) uptake and GLUT4myc translocation. Concomitant overexpression of GLUT4myc and Rab11 wt in cardiomyocytes decreased the amount of GLUT4myc at the cell surface by about 50%, an effect not observed for Rab11 N124I. However, the dominant-negative mutant reduced the efficiency of insulin to promote glucose uptake and GLUT4 translocation in both cardiac and skeletal muscle cells to about one half. The level of Akt phosphorylation does not vary after cotransfection indicating that insulin signalling remained unaffected under these conditions. In conclusion, our data show that Rab11 (i) mediates endocytosis of GLUT4 and (ii) plays a pivotal role in insulin-regulated translocation of this transporter to the plasma membrane.
Collapse
Affiliation(s)
- Mathias Uhlig
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Auf'm Hennekamp 65, D-40225 Düsseldorf, Germany
| | | | | |
Collapse
|
48
|
Zhang X, Carey GB. Plasma membrane-bound cyclic AMP phosphodiesterase activity in 3T3-L1 adipocytes. Comp Biochem Physiol B Biochem Mol Biol 2005; 137:309-16. [PMID: 15050518 DOI: 10.1016/j.cbpc.2003.12.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2003] [Revised: 12/02/2003] [Accepted: 12/07/2003] [Indexed: 11/22/2022]
Abstract
Plasma membranes were isolated from 3T3-L1 adipocytes. Plasma membrane phosphodiesterase (PM-PDE) was measured in the presence of 5 microM cilostamide. Time course and cAMP dose response ranging from 0 to 2 microM were measured. PM-PDE remained linear up to 20 min. Non-linear curve fitting analysis showed that the low Km cAMP dose data fit a two component curve significantly better than a one component curve, indicating that there are two iso-forms of PDE in the plasma membrane of 3T3-L1 adipocytes, similar to swine adipocytes. The Km and Vmax values for this two component curve were Km1=0.12 microM, Vmax1=3.08 pmol min(-1) mg(-1) protein, and Km2=3.67 microM, Vmax2=83.8 pmol min(-1) mg(-1) protein. Inhibitors of PDE1, PDE2 and PDE5 failed to inhibit PM-PDE, as observed in swine adipocyte plasma membranes. However, PDE4 inhibitors were three-fold more effective at inhibiting PDE in 3T3-L1 PM compared to swine adipocyte PM. One mM 1, 3-dipropyl-8-p-sulfophenylxanthine (DPSPX) inhibited PM-PDE by approximately 75% in both preparations. These data demonstrate that PM-PDE is distinct from microsomal membrane PDE and may be responsible for extracellular cAMP metabolism to AMP in 3T3-L1 adipocytes.
Collapse
Affiliation(s)
- Xiujun Zhang
- Department of Animal and Nutritional Sciences, University of New Hampshire, 403 Kendall Hall, Durham, NH 03824, USA
| | | |
Collapse
|
49
|
Proux-Gillardeaux V, Rudge R, Galli T. The Tetanus Neurotoxin-Sensitive and Insensitive Routes to and from the Plasma Membrane: Fast and Slow Pathways? Traffic 2005; 6:366-73. [PMID: 15813747 DOI: 10.1111/j.1600-0854.2005.00288.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Intracellular membrane trafficking in eukaryotes involves the budding of vesicles from a donor compartment, their translocation, and subsequent fusion with a target membrane. This last step has been shown to involve SNARE proteins, classified into two categories, vesicular (v)-SNAREs and target (t)-SNAREs. It is the pairing of v- and t-SNAREs that is responsible for bringing the lipid bilayers together for membrane fusion. Key to the discovery of SNAREs is the sensitivity of their neuronal synaptic prototypes, which mediate the release of neurotransmitters, to clostridial neurotoxins. In this review, we focus on tetanus neurotoxin-sensitive and tetanus neurotoxin-insensitive v-SNAREs, in particular synaptobrevin and cellubrevin, both tetanus neurotoxin-sensitive and Tetanus neurotoxin-Insensitive Vesicle-Associated Membrane Protein (TI-VAMP, also called VAMP7). The brevins are characterized by an RD sequence in the middle of their SNARE motif whereas TI-VAMP has an RG sequence. These two categories of exocytic v-SNAREs define two important routes to and from the plasma membrane: one sensitive, the other insensitive to tetanus neurotoxin. We also discuss the central role of the endosomal system that could be considered, as already suggested for Rab proteins, as a mosaic of v-SNAREs, thus raising the question of whether or not these two routes can merge, and if so, how and where.
Collapse
Affiliation(s)
- Véronique Proux-Gillardeaux
- Team 'Avenir' INSERM Membrane Traffic in Neuronal & Epithelial Morphogenesis Institut Jacques Monod CNRS/Univ. Paris 6/Univ. Paris 7 UMR7592, 2, place Jussieu, F-75251 Paris Cedex 05, France
| | | | | |
Collapse
|
50
|
Collison M, James DJ, Graham D, Holman GD, Connell JMC, Dominiczak AF, Gould GW, Salt IP. Reduced insulin-stimulated GLUT4 bioavailability in stroke-prone spontaneously hypertensive rats. Diabetologia 2005; 48:539-46. [PMID: 15729573 DOI: 10.1007/s00125-005-1674-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2004] [Accepted: 10/29/2004] [Indexed: 10/25/2022]
Abstract
AIMS/HYPOTHESIS Insulin-stimulated glucose transport is impaired in a genetic model of hypertension, the stroke-prone spontaneously hypertensive rat (SHRSP), yet the molecular mechanisms that underlie this defect in the animals remain unclear. METHODS We examined the effects of insulin on the trafficking of the insulin-responsive glucose transporter GLUT4 to the plasma membrane in isolated adipocytes from SHRSP and normotensive control Wistar-Kyoto (WKY) rats. RESULTS Treatment of isolated adipocytes with insulin resulted in trafficking of GLUT4 to the plasma membrane. There was no significant difference in the magnitude of insulin-stimulated GLUT4 trafficking from intracellular membranes to the plasma membrane between strains. In contrast, we demonstrated that there is a significant reduction in GLUT4 accessible to the glucose photolabel Bio-LC-ATB-BGPA at the plasma membrane of SHRSP adipocytes compared with control rats. CONCLUSIONS/INTERPRETATION We propose that a large proportion of GLUT4 translocated to the plasma membrane in response to insulin is not able to bind substrate and catalyse transport in the SHRSP. Therefore, there is a reduction in bioavailable GLUT4 in SHRSP animals that is likely to account, at least in part, for the reduced insulin-stimulated glucose uptake.
Collapse
Affiliation(s)
- M Collison
- The Henry Wellcome Laboratory of Cell Biology, Division of Biochemistry and Molecular Biology, Institute of Biomedical and Life Sciences, Davidson Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | | | | | | | | | | | | | | |
Collapse
|