1
|
Simsek Papur O, Glatz JFC, Luiken JJFP. Protein kinase-D1 and downstream signaling mechanisms involved in GLUT4 translocation in cardiac muscle. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119748. [PMID: 38723678 DOI: 10.1016/j.bbamcr.2024.119748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 02/21/2024] [Accepted: 04/29/2024] [Indexed: 05/20/2024]
Abstract
The Ser/Thr kinase protein kinase-D1 (PKD1) is involved in induction of various cell physiological processes in the heart such as myocellular hypertrophy and inflammation, which may turn maladaptive during long-term stimulation. Of special interest is a key role of PKD1 in the regulation of cardiac substrate metabolism. Glucose and fatty acids are the most important substrates for cardiac energy provision, and the ratio at which they are utilized determines the health status of the heart. Cardiac glucose uptake is mainly regulated by translocation of the glucose transporter GLUT4 from intracellular stores (endosomes) to the sarcolemma, and fatty acid uptake via a parallel translocation of fatty acid transporter CD36 from endosomes to the sarcolemma. PKD1 is involved in the regulation of GLUT4 translocation, but not CD36 translocation, giving it the ability to modulate glucose uptake without affecting fatty acid uptake, thereby altering the cardiac substrate balance. PKD1 would therefore serve as an attractive target to combat cardiac metabolic diseases with a tilted substrate balance, such as diabetic cardiomyopathy. However, PKD1 activation also elicits cardiac hypertrophy and inflammation. Therefore, identification of the events upstream and downstream of PKD1 may provide superior therapeutic targets to alter the cardiac substrate balance. Recent studies have identified the lipid kinase phosphatidylinositol 4-kinase IIIβ (PI4KIIIβ) as signaling hub downstream of PKD1 to selectively stimulate GLUT4-mediated myocardial glucose uptake without inducing hypertrophy. Taken together, the PKD1 signaling pathway serves a pivotal role in cardiac glucose metabolism and is a promising target to selectively modulate glucose uptake in cardiac disease.
Collapse
Affiliation(s)
- Ozlenen Simsek Papur
- Department of Molecular Medicine, Institute of Health Science, Dokuz Eylül University, Izmir, Turkey
| | - Jan F C Glatz
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; Department of Clinical Genetics, Maastricht University Medical Center(+), Maastricht, the Netherlands
| | - Joost J F P Luiken
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; Department of Clinical Genetics, Maastricht University Medical Center(+), Maastricht, the Netherlands.
| |
Collapse
|
2
|
Ray A, Wen J, Yammine L, Culver J, Parida IS, Garren J, Xue L, Hales K, Xiang Q, Birnbaum MJ, Zhang BB, Monetti M, McGraw TE. Regulated dynamic subcellular GLUT4 localization revealed by proximal proteome mapping in human muscle cells. J Cell Sci 2023; 136:jcs261454. [PMID: 38126809 PMCID: PMC10753500 DOI: 10.1242/jcs.261454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
Regulation of glucose transport, which is central for control of whole-body metabolism, is determined by the amount of GLUT4 glucose transporter (also known as SLC2A4) in the plasma membrane (PM) of fat and muscle cells. Physiologic signals [such as activated insulin receptor or AMP-activated protein kinase (AMPK)] increase PM GLUT4. Here, we show that the distribution of GLUT4 between the PM and interior of human muscle cells is dynamically maintained, and that AMPK promotes PM redistribution of GLUT4 by regulating exocytosis and endocytosis. Stimulation of exocytosis by AMPK is mediated by Rab10 and the Rab GTPase-activating protein TBC1D4. APEX2 proximity mapping reveals that GLUT4 traverses both PM-proximal and PM-distal compartments in unstimulated muscle cells, further supporting retention of GLUT4 by a constitutive retrieval mechanism. AMPK-stimulated translocation involves GLUT4 redistribution among the same compartments traversed in unstimulated cells, with a significant recruitment of GLUT4 from the Golgi and trans-Golgi network compartments. Our comprehensive proximal protein mapping provides an integrated, high-density, whole-cell accounting of the localization of GLUT4 at a resolution of ∼20 nm that serves as a structural framework for understanding the molecular mechanisms regulating GLUT4 trafficking downstream of different signaling inputs in a physiologically relevant cell type.
Collapse
Affiliation(s)
- Anuttoma Ray
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jennifer Wen
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10021, USA
| | - Lucie Yammine
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jeff Culver
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development and Medical, Cambridge, MA 02139, USA
| | | | - Jeonifer Garren
- Global Biometrics and Data Management, Global Product Development, Pfizer Inc., Cambridge, MA 02139, USA
| | - Liang Xue
- Early Clinical Development Biomedicine AI, Pfizer Worldwide Research, Development and Medical, Cambridge, MA 02139, USA
| | - Katherine Hales
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development and Medical, Cambridge, MA 02139, USA
| | - Qing Xiang
- Target Sciences, Pfizer Inc., New York, NY 10016, USA
| | - Morris J. Birnbaum
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development and Medical, Cambridge, MA 02139, USA
| | - Bei B. Zhang
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development and Medical, Cambridge, MA 02139, USA
| | - Mara Monetti
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development and Medical, Cambridge, MA 02139, USA
| | - Timothy E. McGraw
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
3
|
Zhang M, Si SJ, Dai WJ, Yang J, Wang Y, Wei XR, Liu S, Xu CY, Zhang C, Jin F, Jiang LQ. A large particle size is required by a nano/micron sized-fluticasone propionate inhalable suspension for asthma treatment. Colloids Surf B Biointerfaces 2023; 228:113364. [PMID: 37290201 DOI: 10.1016/j.colsurfb.2023.113364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/21/2023] [Accepted: 05/22/2023] [Indexed: 06/10/2023]
Abstract
The nano/micron sized-fluticasone propionate inhalable suspension (FPs) is used for asthma treatment, and this study aimed to elucidate the effects of particle size on the absorption of FPs by various pulmonary cells and the subsequent therapeutic efficacy for asthma. FPs of 727, 1136 and 1612 nm were prepared, and an increase in diameter diminished the endocytosis and macropinocytosis of FPs by alveolar epithelial cells (A549 and Calu-3 cells) but facilitated their uptake by M2-like macrophages; results about the transport across Calu-3 monolayer showed the mucus layer was the main rate-limiting step for the uptake of FPs by epithelial cells; the animal tests showed that although a decrease in diameter improved the pulmonary absorption of FPs, the particle size did not affect the lung distribution of FPs; a further detection revealed that larger FPs were taken more effectively by alveolar macrophages and lymphocytes and exerted a better therapeutic effect on asthma than the smaller ones. This study showed that the particle size of FPs had a significant impact on their absorption, elimination and cellular distribution in the lung after inhalation and further on their effectiveness in asthma treatment, and the particle size of the nano/micron sized-FPs should be designed and optimized for asthma treatment on the premise of meeting the requirements of inhalation preparations.
Collapse
Affiliation(s)
- Mi Zhang
- School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Su Jia Si
- School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Wen Jin Dai
- School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Jian Yang
- The Affiliated Lianyungang Hospital of Xuzhou Medical University, 222002, China
| | - Yan Wang
- School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Xiang Rong Wei
- School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Shuo Liu
- School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Cheng Yi Xu
- School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Cong Zhang
- Shanghai Fangyu Health Pharmaceutical Technology Co., Ltd, Shanghai 200000, China
| | - Fang Jin
- School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| | - Li Qun Jiang
- School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| |
Collapse
|
4
|
Masson SWC, Madsen S, Cooke KC, Potter M, Vegas AD, Carroll L, Thillainadesan S, Cutler HB, Walder KR, Cooney GJ, Morahan G, Stöckli J, James DE. Leveraging genetic diversity to identify small molecules that reverse mouse skeletal muscle insulin resistance. eLife 2023; 12:RP86961. [PMID: 37494090 PMCID: PMC10371229 DOI: 10.7554/elife.86961] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023] Open
Abstract
Systems genetics has begun to tackle the complexity of insulin resistance by capitalising on computational advances to study high-diversity populations. 'Diversity Outbred in Australia (DOz)' is a population of genetically unique mice with profound metabolic heterogeneity. We leveraged this variance to explore skeletal muscle's contribution to whole-body insulin action through metabolic phenotyping and skeletal muscle proteomics of 215 DOz mice. Linear modelling identified 553 proteins that associated with whole-body insulin sensitivity (Matsuda Index) including regulators of endocytosis and muscle proteostasis. To enrich for causality, we refined this network by focusing on negatively associated, genetically regulated proteins, resulting in a 76-protein fingerprint of insulin resistance. We sought to perturb this network and restore insulin action with small molecules by integrating the Broad Institute Connectivity Map platform and in vitro assays of insulin action using the Prestwick chemical library. These complementary approaches identified the antibiotic thiostrepton as an insulin resistance reversal agent. Subsequent validation in ex vivo insulin-resistant mouse muscle and palmitate-induced insulin-resistant myotubes demonstrated potent insulin action restoration, potentially via upregulation of glycolysis. This work demonstrates the value of a drug-centric framework to validate systems-level analysis by identifying potential therapeutics for insulin resistance.
Collapse
Affiliation(s)
- Stewart WC Masson
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
| | - Søren Madsen
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
| | - Kristen C Cooke
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
| | - Meg Potter
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
| | - Alexis Diaz Vegas
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
| | - Luke Carroll
- Australian Proteome Analysis Facility, Macquarie UniversityMacquarie ParkAustralia
| | - Senthil Thillainadesan
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
| | - Harry B Cutler
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
| | - Ken R Walder
- School of Medicine, Deakin UniversityGeelongAustralia
| | - Gregory J Cooney
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
| | - Grant Morahan
- Centre for Diabetes Research, Harry Perkins Institute of Medical ResearchMurdochAustralia
| | - Jacqueline Stöckli
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
- School of Medical Sciences University of SydneySydneyAustralia
| |
Collapse
|
5
|
Ray A, Wen J, Yammine L, Culver J, Garren J, Xue L, Hales K, Xiang Q, Birnbaum MJ, Zhang BB, Monetti M, McGraw TE. GLUT4 dynamic subcellular localization is controlled by AMP kinase activation as revealed by proximal proteome mapping in human muscle cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.543897. [PMID: 37333333 PMCID: PMC10274730 DOI: 10.1101/2023.06.06.543897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Regulation of glucose transport into muscle and adipocytes, central for control of whole-body metabolism, is determined by the amount of GLUT4 glucose transporter in the plasma membrane ( PM ). Physiologic signals (activated insulin receptor or AMP kinase [ AMPK ]), acutely increase PM GLUT4 to enhance glucose uptake. Here we show in kinetic studies that intracellular GLUT4 is in equilibrium with the PM in unstimulated cultured human skeletal muscle cells, and that AMPK promotes GLUT4 redistribution to the PM by regulating both exocytosis and endocytosis. AMPK-stimulation of exocytosis requires Rab10 and Rab GTPase activating protein TBC1D4, requirements shared with insulin control of GLUT4 in adipocytes. Using APEX2 proximity mapping, we identify, at high-density and high-resolution, the GLUT4 proximal proteome, revealing GLUT4 traverses both PM proximal and distal compartments in unstimulated muscle cells. These data support intracellular retention of GLUT4 in unstimulated muscle cells by a dynamic mechanism dependent on the rates of internalization and recycling. AMPK promoted GLUT4 translocation to the PM involves redistribution of GLUT4 among the same compartments traversed in unstimulated cells, with a significant redistribution of GLUT4 from the PM distal Trans Golgi Network Golgi compartments. The comprehensive proximal protein mapping provides an integrated, whole cell accounting of GLUT4's localization at a resolution of ∼20 nm, a structural framework for understanding the molecular mechanisms regulating GLUT4 trafficking downstream of different signaling inputs in physiologically relevant cell type and as such, sheds new light on novel key pathways and molecular components as potential therapeutic approaches to modulate muscle glucose uptake.
Collapse
|
6
|
Rahmani S, Ahmed H, Ibazebo O, Fussner-Dupas E, Wakarchuk WW, Antonescu CN. O-GlcNAc transferase modulates the cellular endocytosis machinery by controlling the formation of clathrin-coated pits. J Biol Chem 2023; 299:102963. [PMID: 36731797 PMCID: PMC9999237 DOI: 10.1016/j.jbc.2023.102963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 02/01/2023] Open
Abstract
Clathrin-mediated endocytosis (CME) controls the internalization and function of a wide range of cell surface proteins. CME occurs by the assembly of clathrin and many other proteins on the inner leaflet of the plasma membrane into clathrin-coated pits (CCPs). These structures recruit specific cargo destined for internalization, generate membrane curvature, and in many cases undergo scission from the plasma membrane to yield intracellular vesicles. The diversity of functions of cell surface proteins controlled via internalization by CME may suggest that regulation of CCP formation could be effective to allow cellular adaptation under different contexts. Of interest is how cues derived from cellular metabolism may regulate CME, given the reciprocal role of CME in controlling cellular metabolism. The modification of proteins with O-linked β-GlcNAc (O-GlcNAc) is sensitive to nutrient availability and may allow cellular adaptation to different metabolic conditions. Here, we examined how the modification of proteins with O-GlcNAc may control CCP formation and thus CME. We used perturbation of key enzymes responsible for protein O-GlcNAc modification, as well as specific mutants of the endocytic regulator AAK1 predicted to be impaired for O-GlcNAc modification. We identify that CCP initiation and the assembly of clathrin and other proteins within CCPs are controlled by O-GlcNAc protein modification. This reveals a new dimension of regulation of CME and highlights the important reciprocal regulation of cellular metabolism and endocytosis.
Collapse
Affiliation(s)
- Sadia Rahmani
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada; Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Hafsa Ahmed
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Osemudiamen Ibazebo
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Eden Fussner-Dupas
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada; Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Warren W Wakarchuk
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.
| | - Costin N Antonescu
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada; Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario, Canada.
| |
Collapse
|
7
|
Bayonés L, Guerra-Fernández MJ, Hinostroza F, Báez-Matus X, Vásquez-Navarrete J, Gallo LI, Parra S, Martínez AD, González-Jamett A, Marengo FD, Cárdenas AM. Gain-of-Function Dynamin-2 Mutations Linked to Centronuclear Myopathy Impair Ca2+-Induced Exocytosis in Human Myoblasts. Int J Mol Sci 2022; 23:ijms231810363. [PMID: 36142275 PMCID: PMC9499313 DOI: 10.3390/ijms231810363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/26/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Gain-of-function mutations of dynamin-2, a mechano-GTPase that remodels membrane and actin filaments, cause centronuclear myopathy (CNM), a congenital disease that mainly affects skeletal muscle tissue. Among these mutations, the variants p.A618T and p.S619L lead to a gain of function and cause a severe neonatal phenotype. By using total internal reflection fluorescence microscopy (TIRFM) in immortalized human myoblasts expressing the pH-sensitive fluorescent protein (pHluorin) fused to the insulin-responsive aminopeptidase IRAP as a reporter of the GLUT4 vesicle trafficking, we measured single pHluorin signals to investigate how p.A618T and p.S619L mutations influence exocytosis. We show here that both dynamin-2 mutations significantly reduced the number and durations of pHluorin signals induced by 10 μM ionomycin, indicating that in addition to impairing exocytosis, they also affect the fusion pore dynamics. These mutations also disrupt the formation of actin filaments, a process that reportedly favors exocytosis. This altered exocytosis might importantly disturb the plasmalemma expression of functional proteins such as the glucose transporter GLUT4 in skeletal muscle cells, impacting the physiology of the skeletal muscle tissue and contributing to the CNM disease.
Collapse
Affiliation(s)
- Lucas Bayonés
- Instituto de Fisiología, Biología Molecular y Neurociencias, CONICET, Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires 1428, Argentina
| | - María José Guerra-Fernández
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Gran Bretaña 1111, Valparaíso 2360102, Chile
| | - Fernando Hinostroza
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, Talca 3460000, Chile
- Centro de Investigación en Neuropsicología y Neurociencias Cognitivas (CINPSI Neurocog), Facultad de Ciencias de la Salud, Universidad Católica del Maule, Talca 3460000, Chile
| | - Ximena Báez-Matus
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Gran Bretaña 1111, Valparaíso 2360102, Chile
| | - Jacqueline Vásquez-Navarrete
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Gran Bretaña 1111, Valparaíso 2360102, Chile
| | - Luciana I. Gallo
- Instituto de Fisiología, Biología Molecular y Neurociencias, CONICET, Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires 1428, Argentina
| | - Sergio Parra
- Instituto de Fisiología Celular—Neurociencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Agustín D. Martínez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Gran Bretaña 1111, Valparaíso 2360102, Chile
| | - Arlek González-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Gran Bretaña 1111, Valparaíso 2360102, Chile
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Fernando D. Marengo
- Instituto de Fisiología, Biología Molecular y Neurociencias, CONICET, Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires 1428, Argentina
- Correspondence: (F.D.M.); (A.M.C.)
| | - Ana M. Cárdenas
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Gran Bretaña 1111, Valparaíso 2360102, Chile
- Correspondence: (F.D.M.); (A.M.C.)
| |
Collapse
|
8
|
Fujise K, Noguchi S, Takeda T. Centronuclear Myopathy Caused by Defective Membrane Remodelling of Dynamin 2 and BIN1 Variants. Int J Mol Sci 2022; 23:ijms23116274. [PMID: 35682949 PMCID: PMC9181712 DOI: 10.3390/ijms23116274] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 02/01/2023] Open
Abstract
Centronuclear myopathy (CNM) is a congenital myopathy characterised by centralised nuclei in skeletal myofibers. T-tubules, sarcolemmal invaginations required for excitation-contraction coupling, are disorganised in the skeletal muscles of CNM patients. Previous studies showed that various endocytic proteins are involved in T-tubule biogenesis and their dysfunction is tightly associated with CNM pathogenesis. DNM2 and BIN1 are two causative genes for CNM that encode essential membrane remodelling proteins in endocytosis, dynamin 2 and BIN1, respectively. In this review, we overview the functions of dynamin 2 and BIN1 in T-tubule biogenesis and discuss how their dysfunction in membrane remodelling leads to CNM pathogenesis.
Collapse
Affiliation(s)
- Kenshiro Fujise
- Departments of Neuroscience and Cell Biology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520-8001, USA;
| | - Satoru Noguchi
- National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo 187-8502, Japan;
| | - Tetsuya Takeda
- Department of Biochemistry, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata-cho 2-5-1, Kita-ku, Okayama 700-8558, Japan
- Correspondence: ; Tel.: +81-86-235-7125; Fax: +81-86-235-7126
| |
Collapse
|
9
|
Ramos PA, Lytle KA, Delivanis D, Nielsen S, LeBrasseur NK, Jensen MD. Insulin-Stimulated Muscle Glucose Uptake and Insulin Signaling in Lean and Obese Humans. J Clin Endocrinol Metab 2021; 106:e1631-e1646. [PMID: 33382888 PMCID: PMC7993573 DOI: 10.1210/clinem/dgaa919] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Indexed: 12/29/2022]
Abstract
PURPOSE Skeletal muscle is the primary site for insulin-stimulated glucose disposal, and muscle insulin resistance is central to abnormal glucose metabolism in obesity. Whether muscle insulin signaling to the level of Akt/AS160 is intact in insulin-resistant obese humans is controversial. METHODS We defined a linear range of insulin-stimulated systemic and leg glucose uptake in 14 obese and 14 nonobese volunteers using a 2-step insulin clamp (Protocol 1) and then examined the obesity-related defects in muscle insulin action in 16 nonobese and 25 obese male and female volunteers matched for fitness using a 1-step, hyperinsulinemic, euglycemic clamp coupled with muscle biopsies (Protocol 2). RESULTS Insulin-stimulated glucose disposal (Si) was reduced by > 60% (P < 0.0001) in the obese group in Protocol 2; however, the phosphorylation of Akt and its downstream effector AS160 were not different between nonobese and obese groups. The increase in phosphorylation of Akt2 in response to insulin was positively correlated with Si for both the nonobese (r = 0.53, P = 0.03) and the obese (r = 0.55, P = 0.01) groups. Total muscle GLUT4 protein was 17% less (P < 0.05) in obese subjects. CONCLUSIONS We suggest that reduced muscle glucose uptake in obesity is not due to defects in the insulin signaling pathway at the level of Akt/AS160, which suggests there remain significant gaps in our knowledge of muscle insulin resistance in obesity. Our data imply that models of acute lipotoxicity do not replicate the pathophysiology of obesity.
Collapse
Affiliation(s)
- Paola A Ramos
- Endocrine Research Unit, Mayo Clinic, Rochester, MN, USA
| | - Kelli A Lytle
- Endocrine Research Unit, Mayo Clinic, Rochester, MN, USA
| | | | - Søren Nielsen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus C, Denmark
| | | | - Michael D Jensen
- Endocrine Research Unit, Mayo Clinic, Rochester, MN, USA
- Correspondence: Michael D. Jensen, MD, Division of Endocrinology, Mayo Clinic, 200 First St SW, Joseph Rm 5–194, Rochester MN 55905, USA.
| |
Collapse
|
10
|
Prunonosa Cervera I, Gabriel BM, Aldiss P, Morton NM. The phospholipase A2 family's role in metabolic diseases: Focus on skeletal muscle. Physiol Rep 2021; 9:e14662. [PMID: 33433056 PMCID: PMC7802192 DOI: 10.14814/phy2.14662] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/17/2022] Open
Abstract
The prevalence of obesity and type 2 diabetes has increased substantially in recent years creating a global health burden. In obesity, skeletal muscle, the main tissue responsible for insulin-mediated glucose uptake, exhibits dysregulation of insulin signaling, glucose uptake, lipid metabolism, and mitochondrial function, thus, promoting type 2 diabetes. The phospholipase A2 (PLA2) enzyme family mediates lipid signaling and membrane remodeling and may play an important role in metabolic disorders such as obesity, diabetes, hyperlipidemia, and fatty liver disease. The PLA2 family consists of 16 members clustered in four groups. PLA2s hydrolyze the sn-2 ester bond of phospholipids generating free fatty acids and lysophospholipids. Differential tissue and subcellular PLA2 expression patterns and the abundance of distinct fatty acyl groups in the target phospholipid determine the impact of individual family members on metabolic functions and, potentially, diseases. Here, we update the current knowledge of the role of the PLA2 family in skeletal muscle, with a view to their potential for therapeutic targeting in metabolic diseases.
Collapse
Affiliation(s)
- Iris Prunonosa Cervera
- Molecular Metabolism GroupCentre for Cardiovascular SciencesQueens Medical Research InstituteUniversity of EdinburghEdinburghUK
| | - Brendan M. Gabriel
- Molecular Metabolism GroupCentre for Cardiovascular SciencesQueens Medical Research InstituteUniversity of EdinburghEdinburghUK
- Department of Physiology and PharmacologyIntegrative PhysiologyKarolinska InstituteStockholmSweden
- Aberdeen Cardiovascular & Diabetes CentreThe Rowett InstituteUniversity of AberdeenAberdeenUK
| | - Peter Aldiss
- Molecular Metabolism GroupCentre for Cardiovascular SciencesQueens Medical Research InstituteUniversity of EdinburghEdinburghUK
| | - Nicholas M. Morton
- Molecular Metabolism GroupCentre for Cardiovascular SciencesQueens Medical Research InstituteUniversity of EdinburghEdinburghUK
| |
Collapse
|
11
|
Huber K, Mestres-Arenas A, Fajas L, Leal-Esteban LC. The multifaceted role of cell cycle regulators in the coordination of growth and metabolism. FEBS J 2020; 288:3813-3833. [PMID: 33030287 PMCID: PMC8359344 DOI: 10.1111/febs.15586] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/08/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022]
Abstract
Adapting to changes in nutrient availability and environmental conditions is a fundamental property of cells. This adaptation requires a multi‐directional coordination between metabolism, growth, and the cell cycle regulators (consisting of the family of cyclin‐dependent kinases (CDKs), their regulatory subunits known as cyclins, CDK inhibitors, the retinoblastoma family members, and the E2F transcription factors). Deciphering the mechanisms accountable for this coordination is crucial for understanding various patho‐physiological processes. While it is well established that metabolism and growth affect cell division, this review will focus on recent observations that demonstrate how cell cycle regulators coordinate metabolism, cell cycle progression, and growth. We will discuss how the cell cycle regulators directly regulate metabolic enzymes and pathways and summarize their involvement in the endolysosomal pathway and in the functions and dynamics of mitochondria.
Collapse
Affiliation(s)
- Katharina Huber
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | | | - Lluis Fajas
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | | |
Collapse
|
12
|
Holman GD. Structure, function and regulation of mammalian glucose transporters of the SLC2 family. Pflugers Arch 2020; 472:1155-1175. [PMID: 32591905 PMCID: PMC7462842 DOI: 10.1007/s00424-020-02411-3] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 12/12/2022]
Abstract
The SLC2 genes code for a family of GLUT proteins that are part of the major facilitator superfamily (MFS) of membrane transporters. Crystal structures have recently revealed how the unique protein fold of these proteins enables the catalysis of transport. The proteins have 12 transmembrane spans built from a replicated trimer substructure. This enables 4 trimer substructures to move relative to each other, and thereby alternately opening and closing a cleft to either the internal or the external side of the membrane. The physiological substrate for the GLUTs is usually a hexose but substrates for GLUTs can include urate, dehydro-ascorbate and myo-inositol. The GLUT proteins have varied physiological functions that are related to their principal substrates, the cell type in which the GLUTs are expressed and the extent to which the proteins are associated with subcellular compartments. Some of the GLUT proteins translocate between subcellular compartments and this facilitates the control of their function over long- and short-time scales. The control of GLUT function is necessary for a regulated supply of metabolites (mainly glucose) to tissues. Pathophysiological abnormalities in GLUT proteins are responsible for, or associated with, clinical problems including type 2 diabetes and cancer and a range of tissue disorders, related to tissue-specific GLUT protein profiles. The availability of GLUT crystal structures has facilitated the search for inhibitors and substrates and that are specific for each GLUT and that can be used therapeutically. Recent studies are starting to unravel the drug targetable properties of each of the GLUT proteins.
Collapse
Affiliation(s)
- Geoffrey D Holman
- Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY, UK.
| |
Collapse
|
13
|
Camus SM, Camus MD, Figueras-Novoa C, Boncompain G, Sadacca LA, Esk C, Bigot A, Gould GW, Kioumourtzoglou D, Perez F, Bryant NJ, Mukherjee S, Brodsky FM. CHC22 clathrin mediates traffic from early secretory compartments for human GLUT4 pathway biogenesis. J Cell Biol 2020; 219:133472. [PMID: 31863584 PMCID: PMC7039200 DOI: 10.1083/jcb.201812135] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 08/02/2019] [Accepted: 10/09/2019] [Indexed: 12/29/2022] Open
Abstract
Blood glucose clearance relies on insulin-stimulated exocytosis of glucose transporter 4 (GLUT4) from sites of sequestration in muscle and fat. This work demonstrates that, in humans, CHC22 clathrin controls GLUT4 traffic from the ER-to-Golgi intermediate compartment to sites of sequestration during GLUT4 pathway biogenesis. Glucose transporter 4 (GLUT4) is sequestered inside muscle and fat and then released by vesicle traffic to the cell surface in response to postprandial insulin for blood glucose clearance. Here, we map the biogenesis of this GLUT4 traffic pathway in humans, which involves clathrin isoform CHC22. We observe that GLUT4 transits through the early secretory pathway more slowly than the constitutively secreted GLUT1 transporter and localize CHC22 to the ER-to-Golgi intermediate compartment (ERGIC). CHC22 functions in transport from the ERGIC, as demonstrated by an essential role in forming the replication vacuole of Legionella pneumophila bacteria, which requires ERGIC-derived membrane. CHC22 complexes with ERGIC tether p115, GLUT4, and sortilin, and downregulation of either p115 or CHC22, but not GM130 or sortilin, abrogates insulin-responsive GLUT4 release. This indicates that CHC22 traffic initiates human GLUT4 sequestration from the ERGIC and defines a role for CHC22 in addition to retrograde sorting of GLUT4 after endocytic recapture, enhancing pathways for GLUT4 sequestration in humans relative to mice, which lack CHC22.
Collapse
Affiliation(s)
- Stéphane M Camus
- Department of Bioengineering and Therapeutic Sciences and Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA.,Department of Microbiology and Immunology and the G.W. Hooper Foundation, University of California, San Francisco, San Francisco, CA.,Division of Biosciences, University College London, London, UK
| | - Marine D Camus
- Department of Bioengineering and Therapeutic Sciences and Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA.,Department of Microbiology and Immunology and the G.W. Hooper Foundation, University of California, San Francisco, San Francisco, CA.,Division of Biosciences, University College London, London, UK
| | | | - Gaelle Boncompain
- Institut Curie, PSL Research University, CNRS UMR 144, Paris, France
| | | | - Christopher Esk
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Anne Bigot
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Association Institut de Myologie, UMR S974 Centre for Research in Myology, Paris, France
| | - Gwyn W Gould
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Dimitrios Kioumourtzoglou
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.,Department of Biology and York Biomedical Research Institute, University of York, York, UK
| | - Franck Perez
- Institut Curie, PSL Research University, CNRS UMR 144, Paris, France
| | - Nia J Bryant
- Department of Biology and York Biomedical Research Institute, University of York, York, UK
| | - Shaeri Mukherjee
- Department of Microbiology and Immunology and the G.W. Hooper Foundation, University of California, San Francisco, San Francisco, CA
| | - Frances M Brodsky
- Department of Bioengineering and Therapeutic Sciences and Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA.,Department of Microbiology and Immunology and the G.W. Hooper Foundation, University of California, San Francisco, San Francisco, CA.,Division of Biosciences, University College London, London, UK
| |
Collapse
|
14
|
Abstract
Dynamin is one of the best-studied membrane fission machineries, which mediates endocytic vesicle pinch-off from the plasma membrane. Among the three dynamin isoforms encoded in mammalian genome, dynamin-2 is the ubiquitously expressed isoform and leads to human muscular or neuronal diseases when mutants causing hyperactivity or hypoactivity of its membrane fission activity occur. While transferrin uptake is the most commonly used assay to measure dynamin activity in cultured cells, here we provide two different methods to quantitatively examine the activity of dynamin in myoblasts and myotubes, i.e., Bin1-tubule vesiculation and glucose transporter 4 fractionation assays, respectively. These methods could provide a quantitative measurement of dynamin activity in both differentiated and undifferentiated myoblasts.
Collapse
Affiliation(s)
- Jessica Laiman
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ya-Wen Liu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Center of Precision Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
15
|
Rahmani S, Defferrari MS, Wakarchuk WW, Antonescu CN. Energetic adaptations: Metabolic control of endocytic membrane traffic. Traffic 2019; 20:912-931. [DOI: 10.1111/tra.12705] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 09/11/2019] [Accepted: 10/13/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Sadia Rahmani
- Department of Chemistry and BiologyRyerson University Toronto Ontario Canada
| | | | - Warren W. Wakarchuk
- Department of Chemistry and BiologyRyerson University Toronto Ontario Canada
- Department of Biological SciencesUniversity of Alberta Edmonton Alberta Canada
| | - Costin N. Antonescu
- Department of Chemistry and BiologyRyerson University Toronto Ontario Canada
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital Toronto Ontario Canada
| |
Collapse
|
16
|
Jiang LQ, Wang TY, Wang Y, Wang ZY, Bai YT. Co-disposition of chitosan nanoparticles by multi types of hepatic cells and their subsequent biological elimination: the mechanism and kinetic studies at the cellular and animal levels. Int J Nanomedicine 2019; 14:6035-6060. [PMID: 31534335 PMCID: PMC6681437 DOI: 10.2147/ijn.s208496] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 07/03/2019] [Indexed: 12/12/2022] Open
Abstract
Background: The clearance of nanomaterials (NMs) from the liver is essential for clinical safety, and their hepatic clearance is primarily determined by the co-disposition process of various types of hepatic cells. Studies of this process and the subsequent clearance routes are urgently needed for organic NMs, which are used as drug carriers more commonly than the inorganic ones. Materials and methods: In this study, the co-disposition of chitosan-based nanoparticles (CsNps) by macrophages and hepatocytes at both the cellular and animal levels as well as their subsequent biological elimination were investigated. RAW264.7 and Hepa1-6 cells were used as models of Kupffer cells and hepatocytes, respectively. Results: The cellular studies showed that CsNps released from RAW264.7 cells could enter Hepa1-6 cells through both clathrin- and caveolin-mediated endocytosis. The transport from Kupffer cells to hepatocytes was also studied in mice, and it was observed that most CsNps localized to the hepatocytes after intravenous injection. Following the distribution in hepatocytes, the hepatobiliary-fecal excretion route was shown to be the primary elimination route for CsNps, besides the kidney-urinary excretion route. The elimination of CsNps in mice was a lengthy process, with a half time of about 2 months. Conclusion: The demonstration in this study of the transport of CsNps from macrophages to hepatocytes and the subsequent hepatobiliary-fecal excretion provides basic information for the future development and clinical application of NMs.
Collapse
Affiliation(s)
- Li-Qun Jiang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Ting-Yu Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Yun Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Zi-Yao Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Yu-Ting Bai
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
- School of Stomatology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
17
|
Polianskyte-Prause Z, Tolvanen TA, Lindfors S, Dumont V, Van M, Wang H, Dash SN, Berg M, Naams JB, Hautala LC, Nisen H, Mirtti T, Groop PH, Wähälä K, Tienari J, Lehtonen S. Metformin increases glucose uptake and acts renoprotectively by reducing SHIP2 activity. FASEB J 2019; 33:2858-2869. [PMID: 30321069 PMCID: PMC6338644 DOI: 10.1096/fj.201800529rr] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 09/24/2018] [Indexed: 01/05/2023]
Abstract
Metformin, the first-line drug to treat type 2 diabetes (T2D), inhibits mitochondrial glycerolphosphate dehydrogenase in the liver to suppress gluconeogenesis. However, the direct target and the underlying mechanisms by which metformin increases glucose uptake in peripheral tissues remain uncharacterized. Lipid phosphatase Src homology 2 domain-containing inositol-5-phosphatase 2 (SHIP2) is upregulated in diabetic rodent models and suppresses insulin signaling by reducing Akt activation, leading to insulin resistance and diminished glucose uptake. Here, we demonstrate that metformin directly binds to and reduces the catalytic activity of the recombinant SHIP2 phosphatase domain in vitro. Metformin inhibits SHIP2 in cultured cells and in skeletal muscle and kidney of db/db mice. In SHIP2-overexpressing myotubes, metformin ameliorates reduced glucose uptake by slowing down glucose transporter 4 endocytosis. SHIP2 overexpression reduces Akt activity and enhances podocyte apoptosis, and both are restored to normal levels by metformin. SHIP2 activity is elevated in glomeruli of patients with T2D receiving nonmetformin medication, but not in patients receiving metformin, compared with people without diabetes. Furthermore, podocyte loss in kidneys of metformin-treated T2D patients is reduced compared with patients receiving nonmetformin medication. Our data unravel a novel molecular mechanism by which metformin enhances glucose uptake and acts renoprotectively by reducing SHIP2 activity.-Polianskyte-Prause, Z., Tolvanen, T. A., Lindfors, S., Dumont, V., Van, M., Wang, H., Dash, S. N., Berg, M., Naams, J.-B., Hautala, L. C., Nisen, H., Mirtti, T., Groop, P.-H., Wähälä, K., Tienari, J., Lehtonen, S. Metformin increases glucose uptake and acts renoprotectively by reducing SHIP2 activity.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Gene Expression Regulation, Enzymologic/drug effects
- Humans
- Hypoglycemic Agents/pharmacology
- Kidney Diseases/prevention & control
- Male
- Metformin/pharmacology
- Mice
- Mice, Inbred C57BL
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/antagonists & inhibitors
- Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism
- Podocytes/cytology
- Podocytes/drug effects
- Podocytes/metabolism
- Rats
Collapse
Affiliation(s)
| | | | - Sonja Lindfors
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Vincent Dumont
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Mervi Van
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Hong Wang
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Surjya N. Dash
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Mika Berg
- Department of Chemistry, University of Helsinki, Helsinki, Finland
| | | | - Laura C. Hautala
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Harry Nisen
- Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Tuomas Mirtti
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Research Center, Folkhälsan Institute of Genetics, University of Helsinki, Helsinki, Finland
- Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
- Central Clinical School, Monash University, Melbourne, Victoria, Australia; and
| | - Kristiina Wähälä
- Department of Chemistry, University of Helsinki, Helsinki, Finland
| | - Jukka Tienari
- Department of Pathology, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital, Hyvinkää, Finland
| | - Sanna Lehtonen
- Department of Pathology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
18
|
Lu X, Chan T, Cheng Z, Shams T, Zhu L, Murray M, Zhou F. The 5'-AMP-Activated Protein Kinase Regulates the Function and Expression of Human Organic Anion Transporting Polypeptide 1A2. Mol Pharmacol 2018; 94:1412-1420. [PMID: 30348897 DOI: 10.1124/mol.118.113423] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/28/2018] [Indexed: 12/14/2022] Open
Abstract
The organic anion transporting polypeptides (OATPs) are important membrane proteins that mediate the cellular uptake of drugs and endogenous substances. OATP1A2 is widely distributed in many human tissues that are targeted in drug therapy; defective OATP1A2 leads to altered drug disposition influencing therapeutic outcomes. 5'-AMP-activated protein kinase (AMPK) signaling plays an important role in the pathogenesis of the metabolic syndrome characterized by an increased incidence of type II diabetes and nonalcoholic fatty liver disease. This study investigated the regulatory role of AMPK in OATP1A2 transport function and expression. We found that the treatment of AMPK-specific inhibitor compound C (dorsomorphin dihydrochloride) decreased OATP1A2-mediated uptake of estrone-3-sulfate in a concentration- and time-dependent manner. The impaired OATP1A2 function was associated with a reduced Vmax [154.6 ± 17.9 pmol × (μg × 4 minutes)-1 in compound C-treated cells vs. 413.6 ± 52.5 pmol × (μg × 4 minutes)-1 in controls]; the Km was unchanged. The cell-surface expression of OATP1A2 was decreased by compound C treatment, but total cellular expression was unchanged. The impaired cell-surface expression of OATP1A2 was associated with accelerated internalization and impaired targeting/recycling. Silencing of the AMPK α1-subunit using specific small interfering RNA corroborated the findings with compound C and revealed a role for AMPK in regulating OATP1A2 protein stability. Overall, this study implicated AMPK in the regulation of the function and expression of OATP1A2, which potentially impacts on the disposition of OATP1A2 drug substrates that may be used to treat patients with the metabolic syndrome and other diseases.
Collapse
Affiliation(s)
- Xiaoxi Lu
- School of Pharmacy (X.L., T.C., Z.C., T.S., F.Z.), Save Sight Institute (L.Z.), and Discipline of Pharmacology (M.M.), Faculty of Medicine and Health, University of Sydney, New South Wales, Australia; and Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan (T.C.)
| | - Ting Chan
- School of Pharmacy (X.L., T.C., Z.C., T.S., F.Z.), Save Sight Institute (L.Z.), and Discipline of Pharmacology (M.M.), Faculty of Medicine and Health, University of Sydney, New South Wales, Australia; and Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan (T.C.)
| | - Zhengqi Cheng
- School of Pharmacy (X.L., T.C., Z.C., T.S., F.Z.), Save Sight Institute (L.Z.), and Discipline of Pharmacology (M.M.), Faculty of Medicine and Health, University of Sydney, New South Wales, Australia; and Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan (T.C.)
| | - Tahiatul Shams
- School of Pharmacy (X.L., T.C., Z.C., T.S., F.Z.), Save Sight Institute (L.Z.), and Discipline of Pharmacology (M.M.), Faculty of Medicine and Health, University of Sydney, New South Wales, Australia; and Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan (T.C.)
| | - Ling Zhu
- School of Pharmacy (X.L., T.C., Z.C., T.S., F.Z.), Save Sight Institute (L.Z.), and Discipline of Pharmacology (M.M.), Faculty of Medicine and Health, University of Sydney, New South Wales, Australia; and Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan (T.C.)
| | - Michael Murray
- School of Pharmacy (X.L., T.C., Z.C., T.S., F.Z.), Save Sight Institute (L.Z.), and Discipline of Pharmacology (M.M.), Faculty of Medicine and Health, University of Sydney, New South Wales, Australia; and Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan (T.C.)
| | - Fanfan Zhou
- School of Pharmacy (X.L., T.C., Z.C., T.S., F.Z.), Save Sight Institute (L.Z.), and Discipline of Pharmacology (M.M.), Faculty of Medicine and Health, University of Sydney, New South Wales, Australia; and Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan (T.C.)
| |
Collapse
|
19
|
Hinze C, Boucrot E. Endocytosis in proliferating, quiescent and terminally differentiated cells. J Cell Sci 2018; 131:131/23/jcs216804. [PMID: 30504135 DOI: 10.1242/jcs.216804] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Endocytosis mediates nutrient uptake, receptor internalization and the regulation of cell signaling. It is also hijacked by many bacteria, viruses and toxins to mediate their cellular entry. Several endocytic routes exist in parallel, fulfilling different functions. Most studies on endocytosis have used transformed cells in culture. However, as the majority of cells in an adult body have exited the cell cycle, our understanding is biased towards proliferating cells. Here, we review the evidence for the different pathways of endocytosis not only in dividing, but also in quiescent, senescent and terminally differentiated cells. During mitosis, residual endocytosis is dedicated to the internalization of caveolae and specific receptors. In non-dividing cells, clathrin-mediated endocytosis (CME) functions, but the activity of alternative processes, such as caveolae, macropinocytosis and clathrin-independent routes, vary widely depending on cell types and functions. Endocytosis supports the quiescent state by either upregulating cell cycle arrest pathways or downregulating mitogen-induced signaling, thereby inhibiting cell proliferation. Endocytosis in terminally differentiated cells, such as skeletal muscles, adipocytes, kidney podocytes and neurons, supports tissue-specific functions. Finally, uptake is downregulated in senescent cells, making them insensitive to proliferative stimuli by growth factors. Future studies should reveal the molecular basis for the differences in activities between the different cell states.
Collapse
Affiliation(s)
- Claudia Hinze
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Emmanuel Boucrot
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, UK .,Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, London WC1E 7HX, UK
| |
Collapse
|
20
|
PKC and Rab13 mediate Ca2+ signal-regulated GLUT4 traffic. Biochem Biophys Res Commun 2018; 495:1956-1963. [DOI: 10.1016/j.bbrc.2017.12.064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 12/12/2017] [Indexed: 11/17/2022]
|
21
|
Russell J, Du Toit EF, Peart JN, Patel HH, Headrick JP. Myocyte membrane and microdomain modifications in diabetes: determinants of ischemic tolerance and cardioprotection. Cardiovasc Diabetol 2017; 16:155. [PMID: 29202762 PMCID: PMC5716308 DOI: 10.1186/s12933-017-0638-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/22/2017] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease, predominantly ischemic heart disease (IHD), is the leading cause of death in diabetes mellitus (DM). In addition to eliciting cardiomyopathy, DM induces a ‘wicked triumvirate’: (i) increasing the risk and incidence of IHD and myocardial ischemia; (ii) decreasing myocardial tolerance to ischemia–reperfusion (I–R) injury; and (iii) inhibiting or eliminating responses to cardioprotective stimuli. Changes in ischemic tolerance and cardioprotective signaling may contribute to substantially higher mortality and morbidity following ischemic insult in DM patients. Among the diverse mechanisms implicated in diabetic impairment of ischemic tolerance and cardioprotection, changes in sarcolemmal makeup may play an overarching role and are considered in detail in the current review. Observations predominantly in animal models reveal DM-dependent changes in membrane lipid composition (cholesterol and triglyceride accumulation, fatty acid saturation vs. reduced desaturation, phospholipid remodeling) that contribute to modulation of caveolar domains, gap junctions and T-tubules. These modifications influence sarcolemmal biophysical properties, receptor and phospholipid signaling, ion channel and transporter functions, contributing to contractile and electrophysiological dysfunction, cardiomyopathy, ischemic intolerance and suppression of protective signaling. A better understanding of these sarcolemmal abnormalities in types I and II DM (T1DM, T2DM) can inform approaches to limiting cardiomyopathy, associated IHD and their consequences. Key knowledge gaps include details of sarcolemmal changes in models of T2DM, temporal patterns of lipid, microdomain and T-tubule changes during disease development, and the precise impacts of these diverse sarcolemmal modifications. Importantly, exercise, dietary, pharmacological and gene approaches have potential for improving sarcolemmal makeup, and thus myocyte function and stress-resistance in this ubiquitous metabolic disorder.
Collapse
Affiliation(s)
- Jake Russell
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Eugene F Du Toit
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Jason N Peart
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Hemal H Patel
- VA San Diego Healthcare System and Department of Anesthesiology, University of California San Diego, San Diego, USA
| | - John P Headrick
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia. .,School of Medical Science, Griffith University, Southport, QLD, 4217, Australia.
| |
Collapse
|
22
|
González-Jamett AM, Baez-Matus X, Olivares MJ, Hinostroza F, Guerra-Fernández MJ, Vasquez-Navarrete J, Bui MT, Guicheney P, Romero NB, Bevilacqua JA, Bitoun M, Caviedes P, Cárdenas AM. Dynamin-2 mutations linked to Centronuclear Myopathy impair actin-dependent trafficking in muscle cells. Sci Rep 2017; 7:4580. [PMID: 28676641 PMCID: PMC5496902 DOI: 10.1038/s41598-017-04418-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 05/16/2017] [Indexed: 12/20/2022] Open
Abstract
Dynamin-2 is a ubiquitously expressed GTP-ase that mediates membrane remodeling. Recent findings indicate that dynamin-2 also regulates actin dynamics. Mutations in dynamin-2 cause dominant centronuclear myopathy (CNM), a congenital myopathy characterized by progressive weakness and atrophy of skeletal muscles. However, the muscle-specific roles of dynamin-2 affected by these mutations remain elusive. Here we show that, in muscle cells, the GTP-ase activity of dynamin-2 is involved in de novo actin polymerization as well as in actin-mediated trafficking of the glucose transporter GLUT4. Expression of dynamin-2 constructs carrying CNM-linked mutations disrupted the formation of new actin filaments as well as the stimulus-induced translocation of GLUT4 to the plasma membrane. Similarly, mature muscle fibers isolated from heterozygous knock-in mice that harbor the dynamin-2 mutation p.R465W, an animal model of CNM, exhibited altered actin organization, reduced actin polymerization and impaired insulin-induced translocation of GLUT4 to the sarcolemma. Moreover, GLUT4 displayed aberrant perinuclear accumulation in biopsies from CNM patients carrying dynamin-2 mutations, further suggesting trafficking defects. These results suggest that dynamin-2 is a key regulator of actin dynamics and GLUT4 trafficking in muscle cells. Our findings also support a model in which impairment of actin-dependent trafficking contributes to the pathological mechanism in dynamin-2-associated CNM.
Collapse
Affiliation(s)
- Arlek M González-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile. .,Programa de Farmacología Molecular y Clinica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| | - Ximena Baez-Matus
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - María José Olivares
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Fernando Hinostroza
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.,Doctorado en Ciencias, mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Maria José Guerra-Fernández
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Jacqueline Vasquez-Navarrete
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Mai Thao Bui
- Université Sorbonne, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Paris, France.,Centre de référence de Pathologie Neuromusculaire Paris-Est, Institut de Myologie, GHU Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, GH Pitié-Salpêtrière, Paris, France
| | - Pascale Guicheney
- INSERM, UMR_S1166, Sorbonne Universités, UPMC Univ Paris 06, UMR_S1166, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Norma Beatriz Romero
- Université Sorbonne, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Paris, France.,Centre de référence de Pathologie Neuromusculaire Paris-Est, Institut de Myologie, GHU Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, GH Pitié-Salpêtrière, Paris, France
| | - Jorge A Bevilacqua
- Programa de Anatomía y Biología del Desarrollo, ICBM, Facultad de Medicina, Departamento de Neurología y Neurocirugía, Hospital Clínico Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Marc Bitoun
- Research Center for Myology, UPMC Univ Paris 06 and INSERM UMRS 974, Institute of Myology, Paris, France
| | - Pablo Caviedes
- Programa de Farmacología Molecular y Clinica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| | - Ana M Cárdenas
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
23
|
Barrientos G, Sánchez-Aguilera P, Jaimovich E, Hidalgo C, Llanos P. Membrane Cholesterol in Skeletal Muscle: A Novel Player in Excitation-Contraction Coupling and Insulin Resistance. J Diabetes Res 2017; 2017:3941898. [PMID: 28367451 PMCID: PMC5358446 DOI: 10.1155/2017/3941898] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 02/06/2017] [Indexed: 11/17/2022] Open
Abstract
Membrane cholesterol is critical for signaling processes in a variety of tissues. We will address here current evidence supporting an emerging role of cholesterol on excitation-contraction coupling and glucose transport in skeletal muscle. We have centered our review on the transverse tubule system, a complex network of narrow plasma membrane invaginations that propagate membrane depolarization into the fiber interior and allow nutrient delivery into the fibers. We will discuss current evidence showing that transverse tubule membranes have remarkably high cholesterol levels and we will address how modifications of cholesterol content influence excitation-contraction coupling. In addition, we will discuss how membrane cholesterol levels affect glucose transport by modulating the insertion into the membrane of the main insulin-sensitive glucose transporter GLUT4. Finally, we will address how the increased membrane cholesterol levels displayed by obese animals, which also present insulin resistance, affect these two particular skeletal muscle functions.
Collapse
Affiliation(s)
- G. Barrientos
- Center for Molecular Studies of the Cell, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Physiology and Biophysics Program, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - P. Sánchez-Aguilera
- Center for Molecular Studies of the Cell, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Institute for Research in Dental Sciences, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - E. Jaimovich
- Center for Molecular Studies of the Cell, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Cell and Molecular Biology Program, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - C. Hidalgo
- Center for Molecular Studies of the Cell, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Physiology and Biophysics Program, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- BNI, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - P. Llanos
- Center for Molecular Studies of the Cell, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Institute for Research in Dental Sciences, Facultad de Odontología, Universidad de Chile, Santiago, Chile
- *P. Llanos:
| |
Collapse
|
24
|
Tunduguru R, Thurmond DC. Promoting Glucose Transporter-4 Vesicle Trafficking along Cytoskeletal Tracks: PAK-Ing Them Out. Front Endocrinol (Lausanne) 2017; 8:329. [PMID: 29209279 PMCID: PMC5701999 DOI: 10.3389/fendo.2017.00329] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/06/2017] [Indexed: 12/27/2022] Open
Abstract
Glucose is the principal cellular energy source in humans and maintenance of glucose homeostasis is critical for survival. Glucose uptake into peripheral skeletal muscle and adipose tissues requires the trafficking of vesicles containing glucose transporter-4 (GLUT4) from the intracellular storage compartments to the cell surface. Trafficking of GLUT4 storage vesicles is initiated via the canonical insulin signaling cascade in skeletal muscle and fat cells, as well as via exercise-induced contraction in muscle cells. Recent studies have elucidated steps in the signaling cascades that involve remodeling of the cytoskeleton, a process that underpins the mechanical movement of GLUT4 vesicles. This review is focused upon an alternate phosphoinositide-3 kinase-dependent pathway involving Ras-related C3 botulinum toxin substrate 1 signaling through the p21-activated kinase p21-activated kinase 1 and showcases related signaling events that co-regulate both the depolymerization and re-polymerization of filamentous actin. These new insights provide an enriched understanding into the process of glucose transport and yield potential new targets for interventions aimed to improve insulin sensitivity and remediate insulin resistance, pre-diabetes, and the progression to type 2 diabetes.
Collapse
Affiliation(s)
- Ragadeepthi Tunduguru
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA, United States
| | - Debbie C. Thurmond
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA, United States
- *Correspondence: Debbie C. Thurmond,
| |
Collapse
|
25
|
TUSC5 regulates insulin-mediated adipose tissue glucose uptake by modulation of GLUT4 recycling. Mol Metab 2015; 4:795-810. [PMID: 26629404 PMCID: PMC4632119 DOI: 10.1016/j.molmet.2015.08.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 08/11/2015] [Accepted: 08/18/2015] [Indexed: 12/26/2022] Open
Abstract
Objective Failure to properly dispose of glucose in response to insulin is a serious health problem, occurring during obesity and is associated with type 2 diabetes development. Insulin-stimulated glucose uptake is facilitated by the translocation and plasma membrane fusion of vesicles containing glucose transporter 4 (GLUT4), the rate-limiting step of post-prandial glucose disposal. Methods We analyzed the role of Tusc5 in the regulation of insulin-stimulated Glut4-mediated glucose uptake in vitro and in vivo. Furthermore, we measured Tusc5 expression in two patient cohorts. Results Herein, we report that TUSC5 controls insulin-stimulated glucose uptake in adipocytes, in vitro and in vivo. TUSC5 facilitates the proper recycling of GLUT4 and other key trafficking proteins during prolonged insulin stimulation, thereby enabling proper protein localization and complete vesicle formation, processes that ultimately enable insulin-stimulated glucose uptake. Tusc5 knockout mice exhibit impaired glucose disposal and TUSC5 expression is predictive of glucose tolerance in obese individuals, independent of body weight. Furthermore, we show that TUSC5 is a PPARγ target and in its absence the anti-diabetic effects of TZDs are significantly blunted. Conclusions Collectively, these findings establish TUSC5 as an adipose tissue-specific protein that enables proper protein recycling, linking the ubiquitous vesicle traffic machinery with tissue-specific insulin-mediated glucose uptake into adipose tissue and the maintenance of a healthy metabolic phenotype in mice and humans. Tusc5 regulates glucose uptake in adipose tissue by modulating the GSV recycling machinery. Tusc5 knockout mice develop insulin resistance due to impaired adipose tissue glucose uptake. Rosiglitazone improves glucose homeostasis in part through the induction of Tusc5. Tusc5 is a novel adipose specific adaptor protein linking Glut4 trafficking to the ubiquitous machinery.
Collapse
|
26
|
Garay C, Judge G, Lucarelli S, Bautista S, Pandey R, Singh T, Antonescu CN. Epidermal growth factor-stimulated Akt phosphorylation requires clathrin or ErbB2 but not receptor endocytosis. Mol Biol Cell 2015; 26:3504-19. [PMID: 26246598 PMCID: PMC4591694 DOI: 10.1091/mbc.e14-09-1412] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 07/31/2015] [Indexed: 12/20/2022] Open
Abstract
Upon ligand binding, the epidermal growth factor receptor (EGFR) activates signaling and undergoes endocytosis. EGFR signaling leading to Akt activation is impaired by perturbation of clathrin but not by inhibition of internalization through perturbation of dynamin. Clathrin may thus directly regulate receptor signaling at the cell surface. Epidermal growth factor (EGF) binding to its receptor (EGFR) activates several signaling intermediates, including Akt, leading to control of cell survival and metabolism. Concomitantly, ligand-bound EGFR is incorporated into clathrin-coated pits—membrane structures containing clathrin and other proteins—eventually leading to receptor internalization. Whether clathrin might regulate EGFR signaling at the plasma membrane before vesicle scission is poorly understood. We compared the effect of clathrin perturbation (preventing formation of, or receptor recruitment to, clathrin structures) to that of dynamin2 (allowing formation of clathrin structures but preventing EGFR internalization) under conditions in which EGFR endocytosis is clathrin dependent. Clathrin perturbation by siRNA gene silencing, with the clathrin inhibitor pitstop2, or knocksideways silencing inhibited EGF-simulated Gab1 and Akt phosphorylation in ARPE-19 cells. In contrast, perturbation of dynamin2 with inhibitors or by siRNA gene silencing did not affect EGF-stimulated Gab1 or Akt phosphorylation. EGF stimulation enriched Gab1 and phospho-Gab1 within clathrin structures. ARPE-19 cells have low ErbB2 expression, and overexpression and knockdown experiments revealed that robust ErbB2 expression bypassed the requirement for clathrin for EGF-stimulated Akt phosphorylation. Thus clathrin scaffolds may represent unique plasma membrane signaling microdomains required for signaling by certain receptors, a function that can be separated from vesicle formation.
Collapse
Affiliation(s)
- Camilo Garay
- Department of Chemistry and Biology, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Gurjeet Judge
- Department of Chemistry and Biology, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Stefanie Lucarelli
- Department of Chemistry and Biology, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Stephen Bautista
- Department of Chemistry and Biology, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Rohan Pandey
- Department of Chemistry and Biology, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Tanveer Singh
- Department of Chemistry and Biology, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Costin N Antonescu
- Department of Chemistry and Biology, Ryerson University, Toronto, ON M5B 2K3, Canada
| |
Collapse
|
27
|
Lorenzo DN, Healy JA, Hostettler J, Davis J, Yang J, Wang C, Hohmeier HE, Zhang M, Bennett V. Ankyrin-B metabolic syndrome combines age-dependent adiposity with pancreatic β cell insufficiency. J Clin Invest 2015; 125:3087-102. [PMID: 26168218 DOI: 10.1172/jci81317] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 05/27/2015] [Indexed: 12/22/2022] Open
Abstract
Rare functional variants of ankyrin-B have been implicated in human disease, including hereditary cardiac arrhythmia and type 2 diabetes (T2D). Here, we developed murine models to evaluate the metabolic consequences of these alterations in vivo. Specifically, we generated knockin mice that express either the human ankyrin-B variant R1788W, which is present in 0.3% of North Americans of mixed European descent and is associated with T2D, or L1622I, which is present in 7.5% of African Americans. Young AnkbR1788W/R1788W mice displayed primary pancreatic β cell insufficiency that was characterized by reduced insulin secretion in response to muscarinic agonists, combined with increased peripheral glucose uptake and concomitantly increased plasma membrane localization of glucose transporter 4 (GLUT4) in skeletal muscle and adipocytes. In contrast, older AnkbR1788W/R1788W and AnkbL1622I/L1622I mice developed increased adiposity, a phenotype that was reproduced in cultured adipocytes, and insulin resistance. GLUT4 trafficking was altered in animals expressing mutant forms of ankyrin-B, and we propose that increased cell surface expression of GLUT4 in skeletal muscle and fatty tissue of AnkbR1788W/R1788W mice leads to the observed age-dependent adiposity. Together, our data suggest that ankyrin-B deficiency results in a metabolic syndrome that combines primary pancreatic β cell insufficiency with peripheral insulin resistance and is directly relevant to the nearly one million North Americans bearing the R1788W ankyrin-B variant.
Collapse
|
28
|
Ross E, Ata R, Thavarajah T, Medvedev S, Bowden P, Marshall JG, Antonescu CN. AMP-Activated Protein Kinase Regulates the Cell Surface Proteome and Integrin Membrane Traffic. PLoS One 2015; 10:e0128013. [PMID: 26010094 PMCID: PMC4444004 DOI: 10.1371/journal.pone.0128013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 04/21/2015] [Indexed: 12/11/2022] Open
Abstract
The cell surface proteome controls numerous cellular functions including cell migration and adhesion, intercellular communication and nutrient uptake. Cell surface proteins are controlled by acute changes in protein abundance at the plasma membrane through regulation of endocytosis and recycling (endomembrane traffic). Many cellular signals regulate endomembrane traffic, including metabolic signaling; however, the extent to which the cell surface proteome is controlled by acute regulation of endomembrane traffic under various conditions remains incompletely understood. AMP-activated protein kinase (AMPK) is a key metabolic sensor that is activated upon reduced cellular energy availability. AMPK activation alters the endomembrane traffic of a few specific proteins, as part of an adaptive response to increase energy intake and reduce energy expenditure. How increased AMPK activity during energy stress may globally regulate the cell surface proteome is not well understood. To study how AMPK may regulate the cell surface proteome, we used cell-impermeable biotinylation to selectively purify cell surface proteins under various conditions. Using ESI-MS/MS, we found that acute (90 min) treatment with the AMPK activator A-769662 elicits broad control of the cell surface abundance of diverse proteins. In particular, A-769662 treatment depleted from the cell surface proteins with functions in cell migration and adhesion. To complement our mass spectrometry results, we used other methods to show that A-769662 treatment results in impaired cell migration. Further, A-769662 treatment reduced the cell surface abundance of β1-integrin, a key cell migration protein, and AMPK gene silencing prevented this effect. While the control of the cell surface abundance of various proteins by A-769662 treatment was broad, it was also selective, as this treatment did not change the cell surface abundance of the transferrin receptor. Hence, the cell surface proteome is subject to acute regulation by treatment with A-769662, at least some of which is mediated by the metabolic sensor AMPK.
Collapse
Affiliation(s)
- Eden Ross
- Department of Chemistry and Biology, Ryerson University, 350 Victoria Street, Toronto, Ontario, M5B 2K3, Canada
| | - Rehman Ata
- Department of Chemistry and Biology, Ryerson University, 350 Victoria Street, Toronto, Ontario, M5B 2K3, Canada
| | - Thanusi Thavarajah
- Department of Chemistry and Biology, Ryerson University, 350 Victoria Street, Toronto, Ontario, M5B 2K3, Canada
| | - Sergei Medvedev
- Department of Chemistry and Biology, Ryerson University, 350 Victoria Street, Toronto, Ontario, M5B 2K3, Canada
| | - Peter Bowden
- Department of Chemistry and Biology, Ryerson University, 350 Victoria Street, Toronto, Ontario, M5B 2K3, Canada
| | - John G Marshall
- Department of Chemistry and Biology, Ryerson University, 350 Victoria Street, Toronto, Ontario, M5B 2K3, Canada
| | - Costin N Antonescu
- Department of Chemistry and Biology, Ryerson University, 350 Victoria Street, Toronto, Ontario, M5B 2K3, Canada
| |
Collapse
|
29
|
Jiang L, Zhao X, Zheng C, Li F, Maclean JL, Chen F, Swami A, Qian H, Zhu J, Ge L. The quantitative detection of the uptake and intracellular fate of albumin nanoparticles. RSC Adv 2015. [DOI: 10.1039/c5ra01683e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Little has been investigated about the intracellular fate of organic nanoparticles (NPs), which is important for the safety and drug delivery efficiency of NPs. In this work, the intracellular disassociation and hydrolysis of albumin NPs were detected based on FRET.
Collapse
Affiliation(s)
- Liqun Jiang
- School of Pharmacy
- China Pharmaceutical University
- Nanjing
- P.R. China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy
| | - Xin Zhao
- Center for Biomedical Engineering
- Department of Medicine
- Brigham and Women's Hospital
- Harvard Medical School
- Boston
| | - Chunli Zheng
- School of Pharmacy
- China Pharmaceutical University
- Nanjing
- P.R. China
| | - Fang Li
- School of Pharmacy
- China Pharmaceutical University
- Nanjing
- P.R. China
- School of Pharmacy
| | | | - Fangcheng Chen
- School of Pharmacy
- China Pharmaceutical University
- Nanjing
- P.R. China
| | - Archana Swami
- Department of Anesthesia
- Brigham and Women's Hospital
- Harvard Medical School
- Boston
- USA
| | - Hai Qian
- School of Pharmacy
- China Pharmaceutical University
- Nanjing
- P.R. China
| | - Jiabi Zhu
- School of Pharmacy
- China Pharmaceutical University
- Nanjing
- P.R. China
| | - Liang Ge
- School of Pharmacy
- China Pharmaceutical University
- Nanjing
- P.R. China
| |
Collapse
|
30
|
Li Q, Zhu X, Ishikura S, Zhang D, Gao J, Sun Y, Contreras-Ferrat A, Foley KP, Lavandero S, Yao Z, Bilan PJ, Klip A, Niu W. Ca²⁺ signals promote GLUT4 exocytosis and reduce its endocytosis in muscle cells. Am J Physiol Endocrinol Metab 2014; 307:E209-24. [PMID: 24895284 DOI: 10.1152/ajpendo.00045.2014] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Elevating cytosolic Ca(2+) stimulates glucose uptake in skeletal muscle, but how Ca(2+) affects intracellular traffic of GLUT4 is unknown. In tissue, changes in Ca(2+) leading to contraction preclude analysis of the impact of individual, Ca(2+)-derived signals. In L6 muscle cells stably expressing GLUT4myc, the Ca(2+) ionophore ionomycin raised cytosolic Ca(2+) and caused a gain in cell surface GLUT4myc. Extra- and intracellular Ca(2+) chelators (EGTA, BAPTA-AM) reversed this response. Ionomycin activated calcium calmodulin kinase II (CaMKII), AMPK, and PKCs, but not Akt. Silencing CaMKIIδ or AMPKα1/α2 partly reduced the ionomycin-induced gain in surface GLUT4myc, as did peptidic or small molecule inhibitors of CaMKII (CN21) and AMPK (Compound C). Compared with the conventional isoenzyme PKC inhibitor Gö6976, the conventional plus novel PKC inhibitor Gö6983 lowered the ionomycin-induced gain in cell surface GLUT4myc. Ionomycin stimulated GLUT4myc exocytosis and inhibited its endocytosis in live cells. siRNA-mediated knockdown of CaMKIIδ or AMPKα1/α2 partly reversed ionomycin-induced GLUT4myc exocytosis but did not prevent its reduced endocytosis. Compared with Gö6976, Gö6983 markedly reversed the slowing of GLUT4myc endocytosis triggered by ionomycin. In summary, rapid Ca(2+) influx into muscle cells accelerates GLUT4myc exocytosis while slowing GLUT4myc endocytosis. CaMKIIδ and AMPK stimulate GLUT4myc exocytosis, whereas novel PKCs reduce endocytosis. These results identify how Ca(2+)-activated signals selectively regulate GLUT4 exocytosis and endocytosis in muscle cells.
Collapse
Affiliation(s)
- Q Li
- Department of Immunology, Key Laboratory of Immuno Microenvironment and Disease of the Educational Ministry of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - X Zhu
- Department of Immunology, Key Laboratory of Immuno Microenvironment and Disease of the Educational Ministry of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - S Ishikura
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - D Zhang
- Department of Immunology, Key Laboratory of Immuno Microenvironment and Disease of the Educational Ministry of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - J Gao
- Clinical Laboratory, First Teaching Hospital of Tianjin University of TCM, Tianjin, China; and
| | - Y Sun
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - A Contreras-Ferrat
- Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas/Facultad Medicina; Universidad de Chile; Santiago, Chile
| | - K P Foley
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - S Lavandero
- Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas/Facultad Medicina; Universidad de Chile; Santiago, Chile
| | - Z Yao
- Department of Immunology, Key Laboratory of Immuno Microenvironment and Disease of the Educational Ministry of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - P J Bilan
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - A Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - W Niu
- Department of Immunology, Key Laboratory of Immuno Microenvironment and Disease of the Educational Ministry of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China;
| |
Collapse
|
31
|
Antonescu CN, McGraw TE, Klip A. Reciprocal regulation of endocytosis and metabolism. Cold Spring Harb Perspect Biol 2014; 6:a016964. [PMID: 24984778 DOI: 10.1101/cshperspect.a016964] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The cellular uptake of many nutrients and micronutrients governs both their cellular availability and their systemic homeostasis. The cellular rate of nutrient or ion uptake (e.g., glucose, Fe(3+), K(+)) or efflux (e.g., Na(+)) is governed by a complement of membrane transporters and receptors that show dynamic localization at both the plasma membrane and defined intracellular membrane compartments. Regulation of the rate and mechanism of endocytosis controls the amounts of these proteins on the cell surface, which in many cases determines nutrient uptake or secretion. Moreover, the metabolic action of diverse hormones is initiated upon binding to surface receptors that then undergo regulated endocytosis and show distinct signaling patterns once internalized. Here, we examine how the endocytosis of nutrient transporters and carriers as well as signaling receptors governs cellular metabolism and thereby systemic (whole-body) metabolite homeostasis.
Collapse
Affiliation(s)
- Costin N Antonescu
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario M5B 2K3, Canada
| | - Timothy E McGraw
- Department of Biochemistry, Weill Medical College of Cornell University, New York, New York 10065
| | - Amira Klip
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| |
Collapse
|
32
|
Puig-Oliveras A, Ramayo-Caldas Y, Corominas J, Estellé J, Pérez-Montarelo D, Hudson NJ, Casellas J, Folch JM, Ballester M. Differences in muscle transcriptome among pigs phenotypically extreme for fatty acid composition. PLoS One 2014; 9:e99720. [PMID: 24926690 PMCID: PMC4057286 DOI: 10.1371/journal.pone.0099720] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 05/19/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Besides having an impact on human health, the porcine muscle fatty acid profile determines meat quality and taste. The RNA-Seq technologies allowed us to explore the pig muscle transcriptome with an unprecedented detail. The aim of this study was to identify differentially-expressed genes between two groups of 6 sows belonging to an Iberian × Landrace backcross with extreme phenotypes according to FA profile. RESULTS We sequenced the muscle transcriptome acquiring 787.5 M of 75 bp paired-end reads. About 85.1% of reads were mapped to the reference genome. Of the total reads, 79.1% were located in exons, 6.0% in introns and 14.9% in intergenic regions, indicating expressed regions not annotated in the reference genome. We identified a 34.5% of the intergenic regions as interspersed repetitive regions. We predicted a total of 2,372 putative proteins. Pathway analysis with 131 differentially-expressed genes revealed that the most statistically-significant metabolic pathways were related with lipid metabolism. Moreover, 18 of the differentially-expressed genes were located in genomic regions associated with IMF composition in an independent GWAS study in the same genetic background. Thus, our results indicate that the lipid metabolism of FAs is differently modulated when the FA composition in muscle differs. For instance, a high content of PUFA may reduce FA and glucose uptake resulting in an inhibition of the lipogenesis. These results are consistent with previous studies of our group analysing the liver and the adipose tissue transcriptomes providing a view of each of the main organs involved in lipid metabolism. CONCLUSIONS The results obtained in the muscle transcriptome analysis increase the knowledge of the gene regulation of IMF deposition, FA profile and meat quality, in terms of taste and nutritional value. Besides, our results may be important in terms of human health.
Collapse
Affiliation(s)
- Anna Puig-Oliveras
- Departament de Genètica Animal, Centre de Recerca en Agrigenòmica (CRAG), Bellaterra, Spain
- Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Yuliaxis Ramayo-Caldas
- Génétique Animale et Biologie Intégrative UMR1313 (GABI), Institut National de la Recherche Agronomique (INRA), Jouy-en-Josas, France
| | - Jordi Corominas
- Departament de Genètica Animal, Centre de Recerca en Agrigenòmica (CRAG), Bellaterra, Spain
- Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Jordi Estellé
- Génétique Animale et Biologie Intégrative UMR1313 (GABI), Institut National de la Recherche Agronomique (INRA), Jouy-en-Josas, France
- Génétique Animale et Biologie Intégrative UMR1313 (GABI), AgroParisTech, Jouy-en-Josas, France
- Laboratoire de Radiobiologie et Etude du Génome (LREG), Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Jouy-en-Josas, France
| | - Dafne Pérez-Montarelo
- Departamento de Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - Nicholas J. Hudson
- Computational and Systems Biology, Commonwealth Scientific and Industrial Research Organisation (CSIRO) Animal, Food and Health SciencesQLD, Brisbane, Australia
| | - Joaquim Casellas
- Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- Departament de Genètica i Millora Animal, Institut de Recerca i Tecnologies Agroalimentàries (IRTA), Lleida, Spain
| | - Josep M. Folch
- Departament de Genètica Animal, Centre de Recerca en Agrigenòmica (CRAG), Bellaterra, Spain
- Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Maria Ballester
- Departament de Genètica Animal, Centre de Recerca en Agrigenòmica (CRAG), Bellaterra, Spain
- Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| |
Collapse
|
33
|
Klip A, Sun Y, Chiu TT, Foley KP. Signal transduction meets vesicle traffic: the software and hardware of GLUT4 translocation. Am J Physiol Cell Physiol 2014; 306:C879-86. [DOI: 10.1152/ajpcell.00069.2014] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Skeletal muscle is the major tissue disposing of dietary glucose, a function regulated by insulin-elicited signals that impart mobilization of GLUT4 glucose transporters to the plasma membrane. This phenomenon, also central to adipocyte biology, has been the subject of intense and productive research for decades. We focus on muscle cell studies scrutinizing insulin signals and vesicle traffic in a spatiotemporal manner. Using the analogy of an integrated circuit to approach the intersection between signal transduction and vesicle mobilization, we identify signaling relays (“software”) that engage structural/mechanical elements (“hardware”) to enact the rapid mobilization and incorporation of GLUT4 into the cell surface. We emphasize how insulin signal transduction switches from tyrosine through lipid and serine phosphorylation down to activation of small G proteins of the Rab and Rho families, describe key negative regulation step of Rab GTPases through the GTPase-activating protein activity of the Akt substrate of 160 kDa (AS160), and focus on the mechanical effectors engaged by Rabs 8A and 10 (the molecular motor myosin Va), and the Rho GTPase Rac1 (actin filament branching and severing through Arp2/3 and cofilin). Finally, we illustrate how actin filaments interact with myosin 1c and α-Actinin4 to promote vesicle tethering as preamble to fusion with the membrane.
Collapse
Affiliation(s)
- Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; and
- Department of Biochemistry, The University of Toronto, Ontario, Canada
| | - Yi Sun
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; and
| | - Tim Ting Chiu
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; and
- Department of Biochemistry, The University of Toronto, Ontario, Canada
| | - Kevin P. Foley
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; and
- Department of Biochemistry, The University of Toronto, Ontario, Canada
| |
Collapse
|
34
|
Foley KP, Klip A. Dynamic GLUT4 sorting through a syntaxin-6 compartment in muscle cells is derailed by insulin resistance-causing ceramide. Biol Open 2014; 3:314-25. [PMID: 24705014 PMCID: PMC4021353 DOI: 10.1242/bio.20147898] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
GLUT4 constitutively recycles between the plasma membrane and intracellular depots. Insulin shifts this dynamic equilibrium towards the plasma membrane by recruiting GLUT4 to the plasma membrane from insulin-responsive vesicles. Muscle is the primary site for dietary glucose deposition; however, how GLUT4 sorts into insulin-responsive vesicles, and if and how insulin resistance affects this process, is unknown. In L6 myoblasts stably expressing myc-tagged GLUT4, we analyzed the intracellular itinerary of GLUT4 as it internalizes from the cell surface and examined if such sorting is perturbed by C2-ceramide, a lipid metabolite causing insulin resistance. Surface-labeled GLUT4myc that internalized for 30 min accumulated in a Syntaxin-6 (Stx6)- and Stx16-positive perinuclear sub-compartment devoid of furin or internalized transferrin, and displayed insulin-responsive re-exocytosis. C2-ceramide dispersed the Stx6-positive sub-compartment and prevented insulin-responsive re-exocytosis of internalized GLUT4myc, even under conditions not affecting insulin-stimulated signaling towards Akt. Microtubule disruption with nocodazole prevented pre-internalized GLUT4myc from reaching the Stx6-positive perinuclear sub-compartment and from undergoing insulin-responsive exocytosis. Removing nocodazole allowed both parameters to recover, suggesting that the Stx6-positive perinuclear sub-compartment was required for GLUT4 insulin-responsiveness. Accordingly, Stx6 knockdown inhibited by ∼50% the ability of internalized GLUT4myc to undergo insulin-responsive re-exocytosis without altering its overall perinuclear accumulation. We propose that Stx6 defines the insulin-responsive compartment in muscle cells. Our data are consistent with a model where ceramide could cause insulin resistance by altering intracellular GLUT4 sorting.
Collapse
Affiliation(s)
- Kevin P Foley
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Amira Klip
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
35
|
Zhang WZ. An association of metabolic syndrome constellation with cellular membrane caveolae. PATHOBIOLOGY OF AGING & AGE RELATED DISEASES 2014; 4:23866. [PMID: 24563731 PMCID: PMC3926988 DOI: 10.3402/pba.v4.23866] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 01/21/2014] [Accepted: 01/21/2014] [Indexed: 01/19/2023]
Abstract
Metabolic syndrome (MetS) is a cluster of metabolic abnormalities that can predispose an individual to a greater risk of developing type-2 diabetes and cardiovascular diseases. The cluster includes abdominal obesity, dyslipidemia, hypertension, and hyperglycemia - all of which are risk factors to public health. While searching for a link among the aforementioned malaises, clues have been focused on the cell membrane domain caveolae, wherein the MetS-associated active molecules are colocalized and interacted with to carry out designated biological activities. Caveola disarray could induce all of those individual metabolic abnormalities to be present in animal models and humans, providing a new target for therapeutic strategy in the management of MetS.
Collapse
Affiliation(s)
- Wei-Zheng Zhang
- CMP Laboratory, Port Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
36
|
Mechanisms regulating GLUT4 transcription in skeletal muscle cells are highly conserved across vertebrates. PLoS One 2013; 8:e80628. [PMID: 24260440 PMCID: PMC3832493 DOI: 10.1371/journal.pone.0080628] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 10/03/2013] [Indexed: 12/30/2022] Open
Abstract
The glucose transporter 4 (GLUT4) plays a key role in glucose uptake in insulin target tissues. This transporter has been extensively studied in many species in terms of its function, expression and cellular traffic and complex mechanisms are involved in its regulation at many different levels. However, studies investigating the transcription of the GLUT4 gene and its regulation are scarce. In this study, we have identified the GLUT4 gene in a teleost fish, the Fugu (Takifugu rubripes), and have cloned and characterized a functional promoter of this gene for the first time in a non-mammalian vertebrate. In silico analysis of the Fugu GLUT4 promoter identified potential binding sites for transcription factors such as SP1, C/EBP, MEF2, KLF, SREBP-1c and GC-boxes, as well as a CpG island, but failed to identify a TATA box. In vitro analysis revealed three transcription start sites, with the main residing 307 bp upstream of the ATG codon. Deletion analysis determined that the core promoter was located between nucleotides -132/+94. By transfecting a variety of 5´deletion constructs into L6 muscle cells we have determined that Fugu GLUT4 promoter transcription is regulated by insulin, PG-J2, a PPARγ agonist, and electrical pulse stimulation. Furthermore, our results suggest the implication of motifs such as PPARγ/RXR and HIF-1α in the regulation of Fugu GLUT4 promoter activity by PPARγ and contractile activity, respectively. These data suggest that the characteristics and regulation of the GLUT4 promoter have been remarkably conserved during the evolution from fish to mammals, further evidencing the important role of GLUT4 in metabolic regulation in vertebrates.
Collapse
|
37
|
Abstract
Glucose is an important fuel for contracting muscle, and normal glucose metabolism is vital for health. Glucose enters the muscle cell via facilitated diffusion through the GLUT4 glucose transporter which translocates from intracellular storage depots to the plasma membrane and T-tubules upon muscle contraction. Here we discuss the current understanding of how exercise-induced muscle glucose uptake is regulated. We briefly discuss the role of glucose supply and metabolism and concentrate on GLUT4 translocation and the molecular signaling that sets this in motion during muscle contractions. Contraction-induced molecular signaling is complex and involves a variety of signaling molecules including AMPK, Ca(2+), and NOS in the proximal part of the signaling cascade as well as GTPases, Rab, and SNARE proteins and cytoskeletal components in the distal part. While acute regulation of muscle glucose uptake relies on GLUT4 translocation, glucose uptake also depends on muscle GLUT4 expression which is increased following exercise. AMPK and CaMKII are key signaling kinases that appear to regulate GLUT4 expression via the HDAC4/5-MEF2 axis and MEF2-GEF interactions resulting in nuclear export of HDAC4/5 in turn leading to histone hyperacetylation on the GLUT4 promoter and increased GLUT4 transcription. Exercise training is the most potent stimulus to increase skeletal muscle GLUT4 expression, an effect that may partly contribute to improved insulin action and glucose disposal and enhanced muscle glycogen storage following exercise training in health and disease.
Collapse
Affiliation(s)
- Erik A Richter
- Molecular Physiology Group, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark.
| | | |
Collapse
|
38
|
Yu C, He B, Xiong MH, Zhang H, Yuan L, Ma L, Dai WB, Wang J, Wang XL, Wang XQ, Zhang Q. The effect of hydrophilic and hydrophobic structure of amphiphilic polymeric micelles on their transport in epithelial MDCK cells. Biomaterials 2013; 34:6284-98. [DOI: 10.1016/j.biomaterials.2013.05.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 05/07/2013] [Indexed: 12/16/2022]
|
39
|
Reed SE, Hodgson LR, Song S, May MT, Kelly EE, McCaffrey MW, Mastick CC, Verkade P, Tavaré JM. A role for Rab14 in the endocytic trafficking of GLUT4 in 3T3-L1 adipocytes. J Cell Sci 2013; 126:1931-41. [PMID: 23444368 DOI: 10.1242/jcs.104307] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Insulin enhances the uptake of glucose into adipocytes and muscle cells by promoting the redistribution of the glucose transporter isoform 4 (GLUT4) from intracellular compartments to the cell surface. Rab GTPases regulate the trafficking itinerary of GLUT4 and several have been found on immunopurified GLUT4 vesicles. Specifically, Rab14 has previously been implicated in GLUT4 trafficking in muscle although its role, if any, in adipocytes is poorly understood. Analysis of 3T3-L1 adipocytes using confocal microscopy demonstrated that endogenous GLUT4 and endogenous Rab14 exhibited a partial colocalisation. However, when wild-type Rab14 or a constitutively-active Rab14Q70L mutant were overexpressed in these cells, the colocalisation with both GLUT4 and IRAP became extensive. Interestingly, this colocalisation was restricted to enlarged 'ring-like' vesicular structures (mean diameter 1.3 µm), which were observed in the presence of overexpressed wild-type Rab14 and Rab14Q70L, but not an inactive Rab14S25N mutant. These enlarged vesicles contained markers of early endosomes and were rapidly filled by GLUT4 and transferrin undergoing endocytosis from the plasma membrane. The Rab14Q70L mutant reduced basal and insulin-stimulated cell surface GLUT4 levels, probably by retaining GLUT4 in an insulin-insensitive early endosomal compartment. Furthermore, shRNA-mediated depletion of Rab14 inhibited the transit of GLUT4 through early endosomal compartments towards vesicles and tubules in the perinuclear region. Given the previously reported role of Rab14 in trafficking between endosomes and the Golgi complex, we propose that the primary role of Rab14 in GLUT4 trafficking is to control the transit of internalised GLUT4 from early endosomes into the Golgi complex, rather than direct GLUT4 translocation to the plasma membrane.
Collapse
Affiliation(s)
- Sam E Reed
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Boguslavsky S, Chiu T, Foley KP, Osorio-Fuentealba C, Antonescu CN, Bayer KU, Bilan PJ, Klip A. Myo1c binding to submembrane actin mediates insulin-induced tethering of GLUT4 vesicles. Mol Biol Cell 2012; 23:4065-78. [PMID: 22918957 PMCID: PMC3469521 DOI: 10.1091/mbc.e12-04-0263] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
GLUT4-containing vesicles cycle between the plasma membrane and intracellular compartments. Insulin promotes GLUT4 exocytosis by regulating GLUT4 vesicle arrival at the cell periphery and its subsequent tethering, docking, and fusion with the plasma membrane. The molecular machinery involved in GLUT4 vesicle tethering is unknown. We show here that Myo1c, an actin-based motor protein that associates with membranes and actin filaments, is required for insulin-induced vesicle tethering in muscle cells. Myo1c was found to associate with both mobile and tethered GLUT4 vesicles and to be required for vesicle capture in the total internal reflection fluorescence (TIRF) zone beneath the plasma membrane. Myo1c knockdown or overexpression of an actin binding-deficient Myo1c mutant abolished insulin-induced vesicle immobilization, increased GLUT4 vesicle velocity in the TIRF zone, and prevented their externalization. Conversely, Myo1c overexpression immobilized GLUT4 vesicles in the TIRF zone and promoted insulin-induced GLUT4 exposure to the extracellular milieu. Myo1c also contributed to insulin-dependent actin filament remodeling. Thus we propose that interaction of vesicular Myo1c with cortical actin filaments is required for insulin-mediated tethering of GLUT4 vesicles and for efficient GLUT4 surface delivery in muscle cells.
Collapse
Affiliation(s)
- Shlomit Boguslavsky
- Cell Biology Program, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Lee WL, Klip A. Shuttling glucose across brain microvessels, with a little help from GLUT1 and AMP kinase. Focus on "AMP kinase regulation of sugar transport in brain capillary endothelial cells during acute metabolic stress". Am J Physiol Cell Physiol 2012; 303:C803-5. [PMID: 22814398 DOI: 10.1152/ajpcell.00241.2012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
42
|
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
|
43
|
Habegger KM, Hoffman NJ, Ridenour CM, Brozinick JT, Elmendorf JS. AMPK enhances insulin-stimulated GLUT4 regulation via lowering membrane cholesterol. Endocrinology 2012; 153:2130-41. [PMID: 22434076 PMCID: PMC3339638 DOI: 10.1210/en.2011-2099] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AMP-activated protein kinase (AMPK) enhances glucose transporter GLUT4 regulation. AMPK also suppresses energy-consuming pathways such as cholesterol synthesis. Interestingly, recent in vitro and in vivo data suggest that excess membrane cholesterol impairs GLUT4 regulation. Therefore, this study tested whether a beneficial, GLUT4-regulatory aspect of AMPK stimulation involved cholesterol lowering. Using L6 myotubes stably expressing an exofacial myc-epitope-tagged-GLUT4, AMPK stimulation by 5-aminoimidazole-4-carboxamide-1-β-d-ribonucleoside (AICAR; 45 min, 1 mm) or 2,4-dinitrophenol (DNP; 30 min, 200 μm) increased cell surface GLUT4myc labeling by approximately ≈ 25% (P < 0.05). Insulin (20 min, 100 nm) also increased GLUT4myc labeling by about 50% (P < 0.05), which was further enhanced (≈ 25%, P < 0.05) by AICAR or DNP. Consistent with AMPK-mediated suppression of cholesterol synthesis, AICAR and DNP decreased membrane cholesterol by 20-25% (P < 0.05). Whereas AMPK knockdown prevented the enhanced basal and insulin-stimulated GLUT4myc labeling by AICAR and DNP, cholesterol replenishment only blocked the AMPK-associated enhancement in insulin action. Cells cultured in a hyperinsulinemic milieu, resembling conditions in vivo that promote the progression/worsening of insulin resistance, displayed an increase in membrane cholesterol. This occurred concomitantly with a loss of cortical filamentous actin (F-actin) and defects in GLUT4 regulation by insulin. These derangements were prevented by AMPK stimulation. Examination of skeletal muscle from insulin-resistant Zucker rats revealed a similar elevation in membrane cholesterol and loss of F-actin. Lowering cholesterol to control levels restored F-actin structure and insulin sensitivity. In conclusion, these data suggest a novel aspect of GLUT4 regulation by AMPK involves membrane cholesterol lowering. Moreover, this AMPK-mediated process protected against hyperinsulinemia-induced insulin resistance.
Collapse
Affiliation(s)
- Kirk M Habegger
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | |
Collapse
|
44
|
McDaniel FK, Molden BM, Mohammad S, Baldini G, McPike L, Narducci P, Granell S, Baldini G. Constitutive cholesterol-dependent endocytosis of melanocortin-4 receptor (MC4R) is essential to maintain receptor responsiveness to α-melanocyte-stimulating hormone (α-MSH). J Biol Chem 2012; 287:21873-90. [PMID: 22544740 DOI: 10.1074/jbc.m112.346890] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Melanocortin-4 receptor (MC4R) is a G-protein-coupled receptor expressed in the hypothalamus where it controls feeding behavior. MC4R cycles constitutively and is internalized at the same rate in the presence or absence of stimulation by the agonist, melanocyte-stimulating hormone (α-MSH). This is different from other G-protein-coupled receptors, such as β(2)-adrenergic receptor (β(2)AR), which internalizes more rapidly in response to agonist stimulation. Here, it is found that in immortalized neuronal Neuro2A cells expressing exogenous receptors, constitutive endocytosis of MC4R and agonist-dependent internalization of β(2)AR were equally sensitive to clathrin depletion. Inhibition of MC4R endocytosis by clathrin depletion decreased the number of receptors at the cell surface that were responsive to the agonist, α-MSH, by 75%. Mild membrane cholesterol depletion also inhibited constitutive endocytosis of MC4R by ∼5-fold, while not affecting recycling of MC4R or agonist-dependent internalization of β(2)AR. Reduced cholesterol did not change the MC4R dose-response curve to α-MSH, but it decreased the amount of cAMP generated per receptor number indicating that a population of MC4R at the cell surface becomes nonfunctional. The loss of MC4R function increased over time (25-50%) and was partially reversed by mutations at putative phosphorylation sites (T312A and S329A). This was reproduced in hypothalamic GT1-7 cells expressing endogenous MC4R. The data indicate that constitutive endocytosis of MC4R is clathrin- and cholesterol-dependent. MC4R endocytosis is required to maintain MC4R responsiveness to α-MSH by constantly eliminating from the plasma membrane a pool of receptors modified at Thr-312 and Ser-329 that have to be cycled to the endosomal compartment to regain function.
Collapse
Affiliation(s)
- Faith K McDaniel
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Strålfors P. Caveolins and Caveolae, Roles in Insulin Signalling and Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 729:111-26. [DOI: 10.1007/978-1-4614-1222-9_8] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
46
|
Bourseau-Guilmain E, Griveau A, Benoit JP, Garcion E. The importance of the stem cell marker prominin-1/CD133 in the uptake of transferrin and in iron metabolism in human colon cancer Caco-2 cells. PLoS One 2011; 6:e25515. [PMID: 21966538 PMCID: PMC3180456 DOI: 10.1371/journal.pone.0025515] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 09/07/2011] [Indexed: 12/22/2022] Open
Abstract
As the pentaspan stem cell marker CD133 was shown to bind cholesterol and to localize in plasma membrane protrusions, we investigated a possible function for CD133 in endocytosis. Using the CD133 siRNA knockdown strategy and non-differentiated human colon cancer Caco-2 cells that constitutively over-expressed CD133, we provide for the first time direct evidence for a role of CD133 in the intracellular accumulation of fluorescently labeled extracellular compounds. Assessed using AC133 monoclonal antibody, CD133 knockdown was shown to improve Alexa488-transferrin (Tf) uptake in Caco-2 cells but had no impact on FITC-dextran or FITC-cholera-toxin. Absence of effect of the CD133 knockdown on Tf recycling established a role for CD133 in inhibiting Tf endocytosis rather than in stimulating Tf exocytosis. Use of previously identified inhibitors of known endocytic pathways and the positive impact of CD133 knockdown on cellular uptake of clathrin-endocytosed synthetic lipid nanocapsules supported that CD133 impact on endocytosis was primarily ascribed to the clathrin pathway. Also, cholesterol extraction with methyl-β-cyclodextrine up regulated Tf uptake at greater intensity in the CD133high situation than in the CD133low situation, thus suggesting a role for cholesterol in the inhibitory effect of CD133 on endocytosis. Interestingly, cell treatment with the AC133 antibody down regulated Tf uptake, thus demonstrating that direct extracellular binding to CD133 could affect endocytosis. Moreover, flow cytometry and confocal microscopy established that down regulation of CD133 improved the accessibility to the TfR from the extracellular space, providing a mechanism by which CD133 inhibited Tf uptake. As Tf is involved in supplying iron to the cell, effects of iron supplementation and deprivation on CD133/AC133 expression were investigated. Both demonstrated a dose-dependent down regulation here discussed to the light of transcriptional and post-transciptional effects. Taken together, these data extend our knowledge of the function of CD133 and underline the interest of further exploring the CD133-Tf-iron network.
Collapse
Affiliation(s)
- Erika Bourseau-Guilmain
- Laboratoire d'Ingénierie de la Vectorisation Particulaire, Inserm, UMR-S 646, Université d'Angers, Angers, France
| | - Audrey Griveau
- Laboratoire d'Ingénierie de la Vectorisation Particulaire, Inserm, UMR-S 646, Université d'Angers, Angers, France
| | - Jean-Pierre Benoit
- Laboratoire d'Ingénierie de la Vectorisation Particulaire, Inserm, UMR-S 646, Université d'Angers, Angers, France
| | - Emmanuel Garcion
- Laboratoire d'Ingénierie de la Vectorisation Particulaire, Inserm, UMR-S 646, Université d'Angers, Angers, France
- * E-mail:
| |
Collapse
|
47
|
Stöckli J, Fazakerley DJ, Coster ACF, Holman GD, James DE. Muscling in on GLUT4 kinetics. Commun Integr Biol 2011; 3:260-2. [PMID: 20714409 DOI: 10.4161/cib.3.3.11457] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Accepted: 02/10/2010] [Indexed: 01/14/2023] Open
Abstract
Insulin triggers glucose uptake into muscle and adipose tissue by stimulating the translocation of the glucose transporter glut4 from intracellular vesicles to the plasma membrane (pm). insulin leads to a rapid increase in glut4 at the pm from approximately 5% to 40-50%. this effect is time and dose-dependent, reaching a new steady state after 30 min of insulin stimulation. previous kinetic analyses in adipocytes has revealed that this is regulated by two mechanisms-increasing the amount of glut4 in the endosomal recycling system and increasing the exocytosis rate constant. fazakerley et al.1 focuses on GLUT4 kinetics in the L6 skeletal muscle cell line. Despite displaying a similar redistribution of GLUT4 to the cell surface with insulin to that seen in adipocytes, the mechanism for this effect in L6 cells was completely different. Insulin had a modest effect to increase the amount of GLUT4 in the recycling system with the dominant effect being on reduction of the endocytosis rate constant. Similar findings were observed with AMPK agonists. These studies indicate that different cell types are capable of achieving the same cell biological endpoint but using completely distinct mechanisms.
Collapse
|
48
|
Buchner DA, Yazbek SN, Solinas P, Burrage LC, Morgan MG, Hoppel CL, Nadeau JH. Increased mitochondrial oxidative phosphorylation in the liver is associated with obesity and insulin resistance. Obesity (Silver Spring) 2011; 19:917-24. [PMID: 20885388 PMCID: PMC3749733 DOI: 10.1038/oby.2010.214] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Obesity is the result of excess energy intake relative to expenditure, however little is known about why some individuals are more prone to weight gain than others. Inbred strains of mice also vary in their susceptibility to obesity and therefore represent a valuable model to study the genetics and physiology of weight gain and its co-morbidities such as type 2 diabetes. C57BL/6J mice are susceptible to obesity and insulin resistance when fed an obesogenic diet, whereas A/J mice are resistant despite increased caloric intake. Analysis of B6- and A/J-derived chromosome substitution strains and congenic strains revealed a complex genetic and physiological basis for this phenotype. To improve our understanding of the molecular mechanisms underlying susceptibility to metabolic disease we analyzed global gene expression patterns in 6C1 and 6C2 congenic strains. 6C1 is susceptible whereas 6C2 is resistant to diet-induced obesity. In addition, we demonstrate that 6C1 is glucose intolerant and insulin resistant relative to 6C2. Pathway analysis of global gene expression patterns in muscle, adipose, and liver identified expression level differences between 6C1 and 6C2 in pathways related to basal transcription factors, endocytosis, and mitochondrial oxidative phosphorylation (OxPhos). The OxPhos expression differences were subtle but evident in each complex of the electron transport chain and were associated with a marked increase in mitochondrial oxidative capacity in the livers of the obese strain 6C1 relative to the obesity-resistant strain 6C2. These data suggests the importance of hepatic mitochondrial function in the development of obesity and insulin resistance.
Collapse
Affiliation(s)
- David A Buchner
- Department of Genetics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA.
| | | | | | | | | | | | | |
Collapse
|
49
|
Berenguer M, Zhang J, Bruce MC, Martinez L, Gonzalez T, Gurtovenko AA, Xu T, Le Marchand-Brustel Y, Govers R. Dimethyl sulfoxide enhances GLUT4 translocation through a reduction in GLUT4 endocytosis in insulin-stimulated 3T3-L1 adipocytes. Biochimie 2011; 93:697-709. [DOI: 10.1016/j.biochi.2010.12.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 12/21/2010] [Indexed: 01/14/2023]
|
50
|
Foley K, Boguslavsky S, Klip A. Endocytosis, recycling, and regulated exocytosis of glucose transporter 4. Biochemistry 2011; 50:3048-61. [PMID: 21405107 DOI: 10.1021/bi2000356] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Glucose transporter 4 (GLUT4) is responsible for the uptake of glucose into muscle and adipose tissues. Under resting conditions, GLUT4 is dynamically retained through idle cycling among selective intracellular compartments, from whence it undergoes slow recycling to the plasma membrane (PM). This dynamic retention can be released by command from intracellular signals elicited by insulin and other stimuli, which result in 2-10-fold increases in the surface level of GLUT4. Insulin-derived signals promote translocation of GLUT4 to the PM from a specialized compartment termed GLUT4 storage vesicles (GSV). Much effort has been devoted to the characterization of the intracellular compartments and dynamics of GLUT4 cycling and to the signals by which GLUT4 is sorted into, and recruited from, GSV. This review summarizes our understanding of intracellular GLUT4 traffic during its internalization from the membrane, its slow, constitutive recycling, and its regulated exocytosis in response to insulin. In spite of specific differences in GLUT4 dynamic behavior in adipose and muscle cells, the generalities of its endocytic and exocytic itineraries are consistent and an array of regulatory proteins that regulate each vesicular traffic event emerges from these cell systems.
Collapse
Affiliation(s)
- Kevin Foley
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario M4G 1X8, Canada
| | | | | |
Collapse
|