1
|
Sneyers F, Speelman-Rooms F, Verhelst SHL, Bootman MD, Bultynck G. Cellular effects of BAPTA: Are they only about Ca 2+ chelation? BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119589. [PMID: 37739271 DOI: 10.1016/j.bbamcr.2023.119589] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/06/2023] [Accepted: 09/10/2023] [Indexed: 09/24/2023]
Abstract
Intracellular Ca2+ signals play a vital role in a broad range of cell biological and physiological processes in all eukaryotic cell types. Dysregulation of Ca2+ signaling has been implicated in numerous human diseases. Over the past four decades, the understanding of how cells use Ca2+ as a messenger has flourished, largely because of the development of reporters that enable visualization of Ca2+ signals in different cellular compartments, and tools that can modulate cellular Ca2+ signaling. One such tool that is frequently used is BAPTA; a fast, high-affinity Ca2+-chelating molecule. By making use of a cell-permeable acetoxymethyl ester (AM) variant, BAPTA can be readily loaded into the cytosol of cells (referred to as BAPTAi), where it is trapped and able to buffer changes in cytosolic Ca2+. Due to the ease of loading of the AM version of BAPTA, this reagent has been used in hundreds of studies to probe the role of Ca2+ signaling in specific processes. As such, for decades, researchers have almost universally attributed changes in biological processes caused by BAPTAi to the involvement of Ca2+ signaling. However, BAPTAi has often been used without any form of control, and in many cases has neither been shown to be retained in cells for the duration of experiments nor to buffer any Ca2+ signals. Moreover, increasing evidence points to off-target cellular effects of BAPTA that are clearly not related to Ca2+ chelation. Here, we briefly introduce Ca2+ signaling and the history of Ca2+ chelators and fluorescent Ca2+ indicators. We highlight Ca2+-independent effects of BAPTAi on a broad range of molecular targets and describe some of BAPTAi's impacts on cell functions that occur independently of its Ca2+-chelating properties. Finally, we propose strategies for determining whether Ca2+ chelation, the binding of other metal ions, or off-target interactions with cell components are responsible for BAPTAi's effect on a particular process and suggest some future research directions.
Collapse
Affiliation(s)
- Flore Sneyers
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, B-3000 Leuven, Belgium
| | - Femke Speelman-Rooms
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, B-3000 Leuven, Belgium; KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Chemical Biology, Herestraat 49 box 901b, B-3000 Leuven, Belgium
| | - Steven H L Verhelst
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Chemical Biology, Herestraat 49 box 901b, B-3000 Leuven, Belgium
| | - Martin D Bootman
- The Open University, Cancer Research Group, School of Life, Health and Chemical Sciences, Milton Keynes, UK
| | - Geert Bultynck
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, B-3000 Leuven, Belgium.
| |
Collapse
|
2
|
Zhou T, Song G, Tian D, Liu Q, Shen J, Yang X, Zhao P. Nuciferine relieves type 2 diabetes mellitus via enhancing GLUT4 expression and translocation. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2023.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
3
|
Mäkinen S, Datta N, Rangarajan S, Nguyen YH, Olkkonen VM, Latva-Rasku A, Nuutila P, Laakso M, Koistinen HA. Finnish-specific AKT2 gene variant leads to impaired insulin signalling in myotubes. J Mol Endocrinol 2023; 70:JME-21-0285. [PMID: 36409629 PMCID: PMC9874976 DOI: 10.1530/jme-21-0285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/21/2022] [Indexed: 11/22/2022]
Abstract
Finnish-specific gene variant p.P50T/AKT2 (minor allele frequency (MAF) = 1.1%) is associated with insulin resistance and increased predisposition to type 2 diabetes. Here, we have investigated in vitro the impact of the gene variant on glucose metabolism and intracellular signalling in human primary skeletal muscle cells, which were established from 14 male p.P50T/AKT2 variant carriers and 14 controls. Insulin-stimulated glucose uptake and glucose incorporation into glycogen were detected with 2-[1,2-3H]-deoxy-D-glucose and D-[14C]-glucose, respectively, and the rate of glycolysis was measured with a Seahorse XFe96 analyzer. Insulin signalling was investigated with Western blotting. The binding of variant and control AKT2-PH domains to phosphatidylinositol (3,4,5)-trisphosphate (PI(3,4,5)P3) was assayed using PIP StripsTM Membranes. Protein tyrosine kinase and serine-threonine kinase assays were performed using the PamGene® kinome profiling system. Insulin-stimulated glucose uptake and glycogen synthesis in myotubes in vitro were not significantly affected by the genotype. However, the insulin-stimulated glycolytic rate was impaired in variant myotubes. Western blot analysis showed that insulin-stimulated phosphorylation of AKT-Thr308, AS160-Thr642 and GSK3β-Ser9 was reduced in variant myotubes compared to controls. The binding of variant AKT2-PH domain to PI(3,4,5)P3 was reduced as compared to the control protein. PamGene® kinome profiling revealed multiple differentially phosphorylated kinase substrates, e.g. calmodulin, between the genotypes. Further in silico upstream kinase analysis predicted a large-scale impairment in activities of kinases participating, for example, in intracellular signal transduction, protein translation and cell cycle events. In conclusion, myotubes from p.P50T/AKT2 variant carriers show multiple signalling alterations which may contribute to predisposition to insulin resistance and T2D in the carriers of this signalling variant.
Collapse
Affiliation(s)
- Selina Mäkinen
- Minerva Foundation Institute for Medical Research, Tukholmankatu, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Haartmaninkatu, Helsinki, Finland
| | - Neeta Datta
- Minerva Foundation Institute for Medical Research, Tukholmankatu, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Haartmaninkatu, Helsinki, Finland
| | - Savithri Rangarajan
- Pam Gene International B.V., Wolvenhoek, BJ ´s-Hertogenbosch, The Netherlands
| | - Yen H Nguyen
- Minerva Foundation Institute for Medical Research, Tukholmankatu, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Haartmaninkatu, Helsinki, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Tukholmankatu, Helsinki, Finland
- Department of Anatomy, Faculty of Medicine, Haartmaninkatu, University of Helsinki, Helsinki, Finland
| | - Aino Latva-Rasku
- Turku PET Centre, University of Turku, Kiinamyllynkatu, Turku, Finland
- Turku PET Centre, Turku University Hospital, Kiinamyllynkatu, Turku, Finland
| | - Pirjo Nuutila
- Turku PET Centre, University of Turku, Kiinamyllynkatu, Turku, Finland
- Turku PET Centre, Turku University Hospital, Kiinamyllynkatu, Turku, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Puijonlaaksontie, Kuopio, Finland
| | - Heikki A Koistinen
- Minerva Foundation Institute for Medical Research, Tukholmankatu, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Haartmaninkatu, Helsinki, Finland
- Correspondence should be addressed to H A Koistinen:
| |
Collapse
|
4
|
Glucose Uptake Is Increased by Estradiol Dipropionate in L6 Skeletal Muscle Cells. Pharmaceuticals (Basel) 2022; 16:ph16010025. [PMID: 36678522 PMCID: PMC9866800 DOI: 10.3390/ph16010025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/05/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
GLUT4 is an important glucose transporter, which is closely related to insulin resistance and type 2 diabetes. In this study, we investigated the mechanism of Estradiol Dipropionate (EDP) on uptake of glucose in L6 skeletal muscle cells. In our study, we confirmed that EDP promoted uptake of glucose in L6 skeletal muscle cells in both normal and insulin resistant models. Western blot indicated that EDP accelerated GLUT4 expression and significantly activated AMPK and PKC phosphorylation; the expression of GLUT4 was significantly inhibited by AMPK inhibitor compound C and PKC inhibitor Gö6983, but not by Wortmannin (Akt inhibitor). Meanwhile, EDP boosted GLUT4 expression, and also increased intracellular Ca2+ levels. In the presence of 2 mM, 0 mM extracellular Ca2+ and 0 mM extracellular Ca2+ + BAPTA-AM, the involvement of intracellular Ca2+ levels contribute to EDP-induced GLUT4 expression and fusion with plasma membrane. Therefore, this study investigated whether EDP promoted GLUT4 expression through AMPK and PKC signaling pathways, thereby enhancing GLUT4 uptake of glucose and fusion into plasma membrane in L6 skeletal muscle cells. In addition, both EDP induced GLUT4 translocation and uptake of glucose were Ca2+ dependent. These findings suggested that EDP may be potential drug for the treatment of type 2 diabetes.
Collapse
|
5
|
Martins VF, LaBarge SA, Stanley A, Svensson K, Hung CW, Keinan O, Ciaraldi TP, Banoian D, Park JE, Ha C, Hetrick B, Meyer GA, Philp A, David LL, Henry RR, Aslan JE, Saltiel AR, McCurdy CE, Schenk S. p300 or CBP is required for insulin-stimulated glucose uptake in skeletal muscle and adipocytes. JCI Insight 2021; 7:141344. [PMID: 34813504 PMCID: PMC8765050 DOI: 10.1172/jci.insight.141344] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 11/19/2021] [Indexed: 11/17/2022] Open
Abstract
While current thinking posits that insulin signaling to GLUT4 exocytic translocation and glucose uptake in skeletal muscle and adipocytes is controlled by phosphorylation-based signaling, many proteins in this pathway are acetylated on lysine residues. However, the importance of acetylation and lysine acetyltransferases to insulin-stimulated glucose uptake is incompletely defined. Here, we demonstrate that combined loss of the acetyltransferases E1A binding protein p300 (p300) and cAMP response element binding protein binding protein (CBP) in mouse skeletal muscle causes a complete loss of insulin-stimulated glucose uptake. Similarly, brief (i.e. 1 h) pharmacological inhibition of p300/CBP acetyltransferase activity recapitulates this phenotype in human and rodent myotubes, 3T3-L1 adipocytes, and mouse muscle. Mechanistically, these effects are due to p300/CBP-mediated regulation of GLUT4 exocytic translocation and occurs downstream of Akt signaling. Taken together, we highlight a fundamental role for acetylation and p300/CBP in the direct regulation of insulin-stimulated glucose transport in skeletal muscle and adipocytes.
Collapse
Affiliation(s)
- Vitor F Martins
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, United States of America
| | - Samuel A LaBarge
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, United States of America
| | - Alexandra Stanley
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, United States of America
| | - Kristoffer Svensson
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, United States of America
| | - Chao-Wei Hung
- Department of Medicine, University of California, San Diego, La Jolla, United States of America
| | - Omer Keinan
- Department of Medicine, University of California, San Diego, La Jolla, United States of America
| | - Theodore P Ciaraldi
- Department of Medicine, University of California, San Diego, La Jolla, United States of America
| | - Dion Banoian
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, United States of America
| | - Ji E Park
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, United States of America
| | - Christina Ha
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, United States of America
| | - Byron Hetrick
- Department of Human Physiology, University of Oregon, Eugene, United States of America
| | - Gretchen A Meyer
- Program in Physical Therapy, Washington University in St. Louis, St. Louis, United States of America
| | - Andrew Philp
- Mitochondrial Metabolism and Ageing, Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Larry L David
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, United States of America
| | - Robert R Henry
- Division of Endocrinology & Metabolism, VA San Diego Healthcare System, San Diego, United States of America
| | - Joseph E Aslan
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, United States of America
| | - Alan R Saltiel
- University of California, San Diego, La Jolla, United States of America
| | - Carrie E McCurdy
- Department of Human Physiology, University of Oregon, Eugene, United States of America
| | - Simon Schenk
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, United States of America
| |
Collapse
|
6
|
Sánchez JC, Valencia-Vásquez A, García AM. Role of TRPV4 Channel in Human White Adipocytes Metabolic Activity. Endocrinol Metab (Seoul) 2021; 36:997-1006. [PMID: 34648704 PMCID: PMC8566118 DOI: 10.3803/enm.2021.1167] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/14/2021] [Accepted: 08/19/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Intracellular calcium (Ca2+) homeostasis plays an essential role in adipocyte metabolism and its alteration is associated with obesity and related disorders. Transient receptor potential vanilloid 4 (TRPV4) channels are an important Ca2+ pathway in adipocytes and their activity is regulated by metabolic mediators such as insulin. In this study, we evaluated the role of TRPV4 channels in metabolic activity and adipokine secretion in human white adipocytes. METHODS Human white adipocytes were freshly cultured and the effects of the activation and inhibition of TRPV4 channels on lipolysis, glucose uptake, lactate production, and leptin and adiponectin secretion were evaluated. RESULTS Under basal and isoproterenol-stimulated conditions, TRPV4 activation by GSK1016709A decreased lipolysis whereas HC067047, an antagonist, increased lipolysis. The activation of TRPV4 resulted in increased glucose uptake and lactate production under both basal conditions and insulin-stimulated conditions; in contrast HC067047 decreased both parameters. Leptin production was increased, and adiponectin production was diminished by TRPV4 activation and its inhibition had the opposite effect. CONCLUSION Our results suggested that TRPV4 channels are metabolic mediators involved in proadipogenic processes and glucose metabolism in adipocyte biology. TRPV4 channels could be a potential pharmacological target to treat metabolic disorders.
Collapse
Affiliation(s)
- Julio C Sánchez
- Faculty of Health Sciences, Technological University of Pereira, La Julita, Pereira, Colombia
| | - Aníbal Valencia-Vásquez
- Faculty of Health Sciences, Technological University of Pereira, La Julita, Pereira, Colombia
| | - Andrés M García
- Faculty of Health Sciences, Technological University of Pereira, La Julita, Pereira, Colombia
| |
Collapse
|
7
|
Complexin-2 redistributes to the membrane of muscle cells in response to insulin and contributes to GLUT4 translocation. Biochem J 2021; 478:407-422. [DOI: 10.1042/bcj20200542] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/11/2020] [Accepted: 01/04/2021] [Indexed: 11/17/2022]
Abstract
Insulin stimulates glucose uptake in muscle cells by rapidly redistributing vesicles containing GLUT4 glucose transporters from intracellular compartments to the plasma membrane (PM). GLUT4 vesicle fusion requires the formation of SNARE complexes between vesicular VAMP and PM syntaxin4 and SNAP23. SNARE accessory proteins usually regulate vesicle fusion processes. Complexins aide in neuro-secretory vesicle-membrane fusion by stabilizing trans-SNARE complexes but their participation in GLUT4 vesicle fusion is unknown. We report that complexin-2 is expressed and homogeneously distributed in L6 rat skeletal muscle cells. Upon insulin stimulation, a cohort of complexin-2 redistributes to the PM. Complexin-2 knockdown markedly inhibited GLUT4 translocation without affecting proximal insulin signalling of Akt/PKB phosphorylation and actin fiber remodelling. Similarly, complexin-2 overexpression decreased maximal GLUT4 translocation suggesting that the concentration of complexin-2 is finely tuned to vesicle fusion. These findings reveal an insulin-dependent regulation of GLUT4 insertion into the PM involving complexin-2.
Collapse
|
8
|
Song G, Huang Y, Xiong M, Yang Z, Liu Q, Shen J, Zhao P, Yang X. Aloperine Relieves Type 2 Diabetes Mellitus via Enhancing GLUT4 Expression and Translocation. Front Pharmacol 2021; 11:561956. [PMID: 33568989 PMCID: PMC7868325 DOI: 10.3389/fphar.2020.561956] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 12/04/2020] [Indexed: 12/17/2022] Open
Abstract
Aloperine (ALO), a quinolizidine alkaloid isolated from Sophora alopecuroides L. used in the traditional Uygur medicine, induced a significant increase in cellular glucose uptake of L6 cells, suggesting it has the potential to relieve hyperglycemia. Therefore, we investigated the effects of ALO on type 2 diabetes mellitus (T2DM) through in vitro and in vivo studies. The translocation of glucose transporter 4 (GLUT4) and changes in intracellular Ca2+ levels were real-time monitored in L6 cells using a laser scanning confocal microscope and related protein kinase inhibitors were used to explore the mechanism of action of ALO. Furthermore, high fat diet combined with low-dose streptozotocin (STZ) was used to induce T2DM in rats, and ALO was given to the stomach of T2DM rats for 4 weeks. In vitro results showed that ALO-induced enhancement of GLUT4 expression and translocation were mediated by G protein-PLC-PKC and PI3K/Akt pathways and ALO-enhanced intracellular Ca2+ was involved in activating PKC via G protein-PLC-IP3R-Ca2+ pathway, resulting in promoted GLUT4 plasma membrane fusion and subsequent glucose uptake. ALO treatment effectively ameliorated hyperglycemia, glucose intolerance, insulin resistance and dyslipidemia, alleviated hepatic steatosis, protected pancreatic islet function and activated GLUT4 expression in insulin target tissues of T2DM rats. These findings demonstrated that ALO deserves attention as a potential hypoglycemic agent.
Collapse
Affiliation(s)
- Guanjun Song
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Yun Huang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Mingrui Xiong
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Ziwei Yang
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Qinghua Liu
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Jinhua Shen
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Ping Zhao
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Xinzhou Yang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| |
Collapse
|
9
|
Brown M, Dainty S, Strudwick N, Mihai AD, Watson JN, Dendooven R, Paton AW, Paton JC, Schröder M. Endoplasmic reticulum stress causes insulin resistance by inhibiting delivery of newly synthesized insulin receptors to the cell surface. Mol Biol Cell 2020; 31:2597-2629. [PMID: 32877278 PMCID: PMC7851869 DOI: 10.1091/mbc.e18-01-0013] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 08/21/2020] [Accepted: 08/28/2020] [Indexed: 12/20/2022] Open
Abstract
Accumulation of unfolded proteins in the endoplasmic reticulum (ER) causes ER stress and activates a signaling network known as the unfolded protein response (UPR). Here we characterize how ER stress and the UPR inhibit insulin signaling. We find that ER stress inhibits insulin signaling by depleting the cell surface population of the insulin receptor. ER stress inhibits proteolytic maturation of insulin proreceptors by interfering with transport of newly synthesized insulin proreceptors from the ER to the plasma membrane. Activation of AKT, a major target of the insulin signaling pathway, by a cytosolic, membrane-bound chimera between the AP20187-inducible FV2E dimerization domain and the cytosolic protein tyrosine kinase domain of the insulin receptor was not affected by ER stress. Hence, signaling events in the UPR, such as activation of the JNK mitogen-activated protein (MAP) kinases or the pseudokinase TRB3 by the ER stress sensors IRE1α and PERK, do not contribute to inhibition of signal transduction in the insulin signaling pathway. Indeed, pharmacologic inhibition and genetic ablation of JNKs, as well as silencing of expression of TRB3, did not restore insulin sensitivity or rescue processing of newly synthesized insulin receptors in ER-stressed cells. [Media: see text].
Collapse
Affiliation(s)
- Max Brown
- Department of Biosciences, Durham University, Durham DH1 3LE, United Kingdom
- Biophysical Sciences Institute, Durham University, Durham DH1 3LE, United Kingdom
- North East England Stem Cell Institute (NESCI), Newcastle Upon Tyne NE1 4EP, United Kingdom
| | - Samantha Dainty
- Department of Biosciences, Durham University, Durham DH1 3LE, United Kingdom
- Biophysical Sciences Institute, Durham University, Durham DH1 3LE, United Kingdom
- North East England Stem Cell Institute (NESCI), Newcastle Upon Tyne NE1 4EP, United Kingdom
| | - Natalie Strudwick
- Department of Biosciences, Durham University, Durham DH1 3LE, United Kingdom
- Biophysical Sciences Institute, Durham University, Durham DH1 3LE, United Kingdom
- North East England Stem Cell Institute (NESCI), Newcastle Upon Tyne NE1 4EP, United Kingdom
| | - Adina D. Mihai
- Department of Biosciences, Durham University, Durham DH1 3LE, United Kingdom
- Biophysical Sciences Institute, Durham University, Durham DH1 3LE, United Kingdom
- North East England Stem Cell Institute (NESCI), Newcastle Upon Tyne NE1 4EP, United Kingdom
| | - Jamie N. Watson
- Department of Biosciences, Durham University, Durham DH1 3LE, United Kingdom
- Biophysical Sciences Institute, Durham University, Durham DH1 3LE, United Kingdom
- North East England Stem Cell Institute (NESCI), Newcastle Upon Tyne NE1 4EP, United Kingdom
| | - Robina Dendooven
- Department of Biosciences, Durham University, Durham DH1 3LE, United Kingdom
- Biophysical Sciences Institute, Durham University, Durham DH1 3LE, United Kingdom
- North East England Stem Cell Institute (NESCI), Newcastle Upon Tyne NE1 4EP, United Kingdom
| | - Adrienne W. Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA 5005, Australia
| | - James C. Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA 5005, Australia
| | - Martin Schröder
- Department of Biosciences, Durham University, Durham DH1 3LE, United Kingdom
- Biophysical Sciences Institute, Durham University, Durham DH1 3LE, United Kingdom
- North East England Stem Cell Institute (NESCI), Newcastle Upon Tyne NE1 4EP, United Kingdom
| |
Collapse
|
10
|
Inoue H, Inazu M, Konishi M, Yokoyama U. Functional expression of TRPM7 as a Ca 2+ influx pathway in adipocytes. Physiol Rep 2020; 7:e14272. [PMID: 31650715 PMCID: PMC6813326 DOI: 10.14814/phy2.14272] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 09/20/2019] [Accepted: 10/04/2019] [Indexed: 02/01/2023] Open
Abstract
In adipocytes, intracellular Ca2+ and Mg2+ modulates physiological functions, such as insulin action and the secretion of adipokines. TRPM7 is a Ca2+/Mg2+‐permeable non‐selective cation channel. TRPM7 mRNA is highly expressed in adipose tissue, however, its functional expression in adipocytes remains to be elucidated. In this study, we demonstrated for the first time that TRPM7 was functionally expressed in both freshly isolated white adipocytes and in 3T3‐L1 adipocytes differentiated from a 3T3‐L1 pre‐adipocyte cell line by whole‐cell patch‐clamp recordings. Consistent with known properties of TRPM7 current, the current in adipocytes was activated by the elimination of extracellular divalent cations and the reduction of intracellular free Mg2+ concentrations, and was inhibited by the TRPM7 inhibitors, 2‐aminoethyl diphenylborinate (2‐APB), hydrogen peroxide (H2O2), N‐methyl maleimide (NMM), NS8593, and 2‐amino‐2‐[2‐(4‐octylphenyl)ethyl]‐1,3‐propanediol (FTY720). Treatment with small‐interfering (si) RNA targeting TRPM7 resulted in a reduction in the current to 23 ± 7% of nontargeting siRNA‐treated adipocytes. Moreover a TRPM7 activator, naltriben, increased the TRPM7‐like current and [Ca2+]i in 3T3‐L1 adipocytes but not in TRPM7‐knockdown adipocytes. These findings indicate that TRPM7 is functionally expressed, and plays a role as a Ca2+ influx pathway in adipocytes.
Collapse
Affiliation(s)
- Hana Inoue
- Department of Physiology, Tokyo Medical University, Tokyo, Japan
| | - Masato Inazu
- Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Masato Konishi
- Department of Physiology, Tokyo Medical University, Tokyo, Japan
| | - Utako Yokoyama
- Department of Physiology, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
11
|
Lozano Jiménez YY, Sánchez Mora RM. Canales de calcio como blanco de interés farmacológico. NOVA 2020. [DOI: 10.22490/24629448.3926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Los canales de calcio son proteínas de membrana que constituyen la vía más importante para el ingreso del ion calcio (Ca2+) a la célula. Al abrirse, permiten el ingreso selectivo del ion, iniciando una variedad de procesos como contracción muscular, secreción endocrina y liberación de neurotransmisores, entre otros. Estas proteínas se agrupan en tres categorías de acuerdo con sus propiedades estructurales y funcionales: (i) Canales de Ca2+ operados por interacción receptor-ligando (ROCC), (ii) Canales activados por parámetros físicos (Transient Receptor Potencial, TRP) y (iii) Canales de Calcio dependientes de voltaje (VDCCs), siendo estos últimos los más estudiados debido a su presencia en células excitables. Dada la importancia de Ca2+ en la fisiología celular, los canales de Ca2+ constituyen un punto de acción farmacológica importante para múltiples tratamientos y, por tanto, son objeto de estudio para el desarrollo de nuevos fármacos. El objetivo de esta revisión es explicar la importancia de los canales de Ca2+ desde una proyección farmacológica, a partir de la exploración documental de artículos publicados hasta la fecha teniendo en cuenta temas relacionados con la estructura de los canales Ca2+, sus propiedades biofísicas, localización celular, funcionamiento y su interacción farmacológica.
Collapse
|
12
|
Ji HH, Ostap EM. The regulatory protein 14-3-3β binds to the IQ motifs of myosin-IC independent of phosphorylation. J Biol Chem 2020; 295:3749-3756. [PMID: 31811090 PMCID: PMC7086031 DOI: 10.1074/jbc.ra119.011227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/21/2019] [Indexed: 12/30/2022] Open
Abstract
Myosin-IC (Myo1c) has been proposed to function in delivery of glucose transporter type 4 (GLUT4)-containing vesicles to the plasma membrane in response to insulin stimulation. Current evidence suggests that, upon insulin stimulation, Myo1c is phosphorylated at Ser701, leading to binding of the signaling protein 14-3-3β. Biochemical and functional details of the Myo1c-14-3-3β interaction have yet to be described. Using recombinantly expressed proteins and mass spectrometry-based analyses to monitor Myo1c phosphorylation, along with pulldown, fluorescence binding, and additional biochemical assays, we show here that 14-3-3β is a dimer and, consistent with previous work, that it binds to Myo1c in the presence of calcium. This interaction was associated with dissociation of calmodulin (CaM) from the IQ motif in Myo1c. Surprisingly, we found that 14-3-3β binds to Myo1c independent of Ser701 phosphorylation in vitro Additionally, in contrast to previous reports, we did not observe Myo1c Ser701 phosphorylation by Ca2+/CaM-dependent protein kinase II (CaMKII), although CaMKII phosphorylated four other Myo1c sites. The presence of 14-3-3β had little effect on the actin-activated ATPase or motile activities of Myo1c. Given these results, it is unlikely that 14-3-3β acts as a cargo adaptor for Myo1c-powered transport; rather, we propose that 14-3-3β binds Myo1c in the presence of calcium and stabilizes the calmodulin-dissociated, nonmotile myosin.
Collapse
Affiliation(s)
- Huan-Hong Ji
- Pennsylvania Muscle Institute, Department of Physiology, and Center for Engineering Mechanobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - E Michael Ostap
- Pennsylvania Muscle Institute, Department of Physiology, and Center for Engineering Mechanobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
13
|
Zhao P, Tian D, Song G, Ming Q, Liu J, Shen J, Liu QH, Yang X. Neferine Promotes GLUT4 Expression and Fusion With the Plasma Membrane to Induce Glucose Uptake in L6 Cells. Front Pharmacol 2019; 10:999. [PMID: 31551792 PMCID: PMC6737894 DOI: 10.3389/fphar.2019.00999] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 08/06/2019] [Indexed: 11/21/2022] Open
Abstract
Glucose transporter 4 (GLUT4) is involved in regulating glucose uptake in striated muscle, liver, and adipose tissue. Neferine is a dibenzyl isoquinoline alkaloid derived from dietary lotus seeds and has multiple pharmacological effects. Therefore, this study investigated neferine’s role in glucose translocation to cell surface, glucose uptake, and GLUT4 expression. In our study, neferine upregulated GLUT4 expression, induced GLUT4 plasma membrane fusion, increased intracellular Ca2+, promoted glucose uptake, and alleviated insulin resistance in L6 cells. Furthermore, neferine significantly activated phosphorylation of AMP-activated protein kinase (AMPK) and protein kinase C (PKC). AMPK and PKC inhibitors blocked neferine-induced GLUT4 expression and increased intracellular Ca2+. While neferine-induced GLUT4 expression and intracellular Ca2+ were inhibited by G protein and PLC inhibitors, only intracellular Ca2+ was inhibited by inositol trisphosphate receptor (IP3R) inhibitors. Thus, neferine promoted GLUT4 expression via the G protein-PLC-PKC and AMPK pathways, inducing GLUT4 plasma membrane fusion and subsequent glucose uptake and increasing intracellular Ca2+ through the G protein-PLC-IP3-IP3R pathway. Treatment with 0 mM extracellular Ca2+ + Ca2+ chelator did not inhibit neferine-induced GLUT4 expression but blocked neferine-induced GLUT4 plasma membrane fusion and glucose uptake, suggesting the latter two are Ca2+-dependent. Therefore, we conclude that neferine is a potential treatment for type 2 diabetes.
Collapse
Affiliation(s)
- Ping Zhao
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China.,National Demonstration Center for Experimental Ethnopharmacology Education, South-Central University for Nationalities, Wuhan, China.,Hubei Medical Biology International Science and Technology Cooperation Base, Wuhan, China
| | - Di Tian
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Guanjun Song
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Qian Ming
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Jia Liu
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Jinhua Shen
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China.,Hubei Medical Biology International Science and Technology Cooperation Base, Wuhan, China
| | - Qing-Hua Liu
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China.,Hubei Medical Biology International Science and Technology Cooperation Base, Wuhan, China
| | - Xinzhou Yang
- National Demonstration Center for Experimental Ethnopharmacology Education, South-Central University for Nationalities, Wuhan, China.,School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| |
Collapse
|
14
|
Zhao P, Ming Q, Qiu J, Tian D, Liu J, Shen J, Liu QH, Yang X. Ethanolic Extract of Folium Sennae Mediates the Glucose Uptake of L6 Cells by GLUT4 and Ca 2. Molecules 2018; 23:molecules23112934. [PMID: 30424024 PMCID: PMC6278344 DOI: 10.3390/molecules23112934] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 11/02/2018] [Accepted: 11/08/2018] [Indexed: 02/06/2023] Open
Abstract
In today’s world, diabetes mellitus (DM) is on the rise, especially type 2 diabetes mellitus (T2DM), which is characterized by insulin resistance. T2DM has high morbidity, and therapies with natural products have attracted much attention in the recent past. In this paper, we aimed to study the hypoglycemic effect and the mechanism of an ethanolic extract of Folium Sennae (FSE) on L6 cells. The glucose uptake of L6 cells was investigated using a glucose assay kit. We studied glucose transporter 4 (GLUT4) expression and AMP-activated protein kinase (AMPK), protein kinase B (PKB/Akt), and protein kinase C (PKC) phosphorylation levels using western blot analysis. GLUT4 trafficking and intracellular Ca2+ levels were monitored by laser confocal microscopy in L6 cells stably expressing IRAP-mOrange. GLUT4 fusion with plasma membrane (PM) was observed by myc-GLUT4-mOrange. FSE stimulated glucose uptake; GLUT4 expression and translocation; PM fusion; intracellular Ca2+ elevation; and the phosphorylation of AMPK, Akt, and PKC in L6 cells. GLUT4 translocation was weakened by the AMPK inhibitor compound C, PI3K inhibitor Wortmannin, PKC inhibitor Gö6983, G protein inhibitor PTX/Gallein, and PLC inhibitor U73122. Similarly, in addition to PTX/Gallein and U73122, the IP3R inhibitor 2-APB and a 0 mM Ca2+-EGTA solution partially inhibited the elevation of intracellular Ca2+ levels. BAPTA-AM had a significant inhibitory effect on FSE-mediated GLUT4 activities. In summary, FSE regulates GLUT4 expression and translocation by activating the AMPK, PI3K/Akt, and G protein–PLC–PKC pathways. FSE causes increasing Ca2+ concentration to complete the fusion of GLUT4 vesicles with PM, allowing glucose uptake. Therefore, FSE may be a potential drug for improving T2DM.
Collapse
Affiliation(s)
- Ping Zhao
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China.
- National Demonstration Center for Experimental Ethnopharmacology Education, South-Central University for Nationalities, Wuhan 430074, China.
| | - Qian Ming
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China.
| | - Junying Qiu
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China.
| | - Di Tian
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China.
| | - Jia Liu
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China.
| | - Jinhua Shen
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China.
| | - Qing-Hua Liu
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China.
| | - Xinzhou Yang
- National Demonstration Center for Experimental Ethnopharmacology Education, South-Central University for Nationalities, Wuhan 430074, China.
- School of Pharmaceutical Sciences, South-Central University for Nationalities, 182 Min-Zu Road, Wuhan 430074, China.
| |
Collapse
|
15
|
Dandelion Chloroform Extract Promotes Glucose Uptake via the AMPK/GLUT4 Pathway in L6 Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:1709587. [PMID: 30524480 PMCID: PMC6247471 DOI: 10.1155/2018/1709587] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/02/2018] [Accepted: 09/04/2018] [Indexed: 01/01/2023]
Abstract
The number of patients with type 2 diabetes mellitus (T2DM) is increasing rapidly worldwide. Glucose transporter 4 (GLUT4) is one of the main proteins that transport blood glucose into the cells and is a target in the treatment of T2DM. In this study, we investigated the mechanism of action of dandelion chloroform extract (DCE) on glucose uptake in L6 cells. The glucose consumption of L6 cell culture supernatant was measured by a glucose uptake assay kit, and the dynamic changes of intracellular GLUT4 and calcium (Ca2+) levels were monitored by laser scanning confocal microscopy in L6 cell lines stably expressing IRAP-mOrange. The GLUT4 fusion with the plasma membrane (PM) was traced via myc-GLUT4-mOrange. GLUT4 expression and AMP-activated protein kinase (AMPK), protein kinase B (PKB/Akt), protein kinase C (PKC), and phosphorylation levels were determined by performing western blotting. GLUT4 mRNA expression was detected by real-time PCR. DCE up-regulated GLUT4 expression, promoted GLUT4 translocation and fusion to the membrane eventually leading to glucose uptake, and induced AMPK phosphorylation in L6 cells. The AMPK inhibitory compound C significantly inhibited DCE-induced GLUT4 expression and translocation while no inhibitory effect was observed by the phosphatidylinositol 3-kinase (PI3K) inhibitor Wortmannin and PKC inhibitor Gö6983. These data suggested that DCE promoted GLUT4 expression and transport to the membrane through the AMPK signaling pathway, thereby stimulating GLUT4 fusion with PM to enhance glucose uptake in L6 cells. DCE-induced GLUT4 translocation was also found to be Ca2+-independent. Together, these findings indicate that DCE could be a new hypoglycemic agent for the treatment of T2DM.
Collapse
|
16
|
Sidibeh CO, Pereira MJ, Abalo XM, J Boersma G, Skrtic S, Lundkvist P, Katsogiannos P, Hausch F, Castillejo-López C, Eriksson JW. FKBP5 expression in human adipose tissue: potential role in glucose and lipid metabolism, adipogenesis and type 2 diabetes. Endocrine 2018; 62:116-128. [PMID: 30032404 PMCID: PMC6153563 DOI: 10.1007/s12020-018-1674-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 07/02/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE Here, we explore the involvement of FKBP51 in glucocorticoid-induced insulin resistance (IR) in human subcutaneous adipose tissue (SAT), including its potential role in type 2 diabetes (T2D). Moreover, we assess the metabolic effects of reducing the activity of FKBP51 using the specific inhibitor SAFit1. METHODS Human SAT was obtained by needle biopsies of the lower abdominal region. FKBP5 gene expression was assessed in fresh SAT explants from a cohort of 20 T2D subjects group-wise matched by gender, age and BMI to 20 non-diabetic subjects. In addition, human SAT was obtained from non-diabetic volunteers (20F/9M). SAT was incubated for 24 h with or without the synthetic glucocorticoid dexamethasone and SAFit1. Incubated SAT was used to measure the glucose uptake rate in isolated adipocytes. RESULTS FKBP5 gene expression levels in SAT positively correlated with several indices of IR as well as glucose area under the curve during oral glucose tolerance test (r = 0.33, p < 0.05). FKBP5 gene expression levels tended to be higher in T2D subjects compared to non-diabetic subjects (p = 0.088). Moreover, FKBP5 gene expression levels were found to inversely correlate with lipolytic, lipogenic and adipogenic genes. SAFit1 partly prevented the inhibitory effects of dexamethasone on glucose uptake. CONCLUSIONS FKBP5 gene expression in human SAT tends to be increased in T2D subjects and is related to elevated glucose levels. Moreover, FKBP5 gene expression is inversely associated with the expression of lipolytic, lipogenic and adipogenic genes. SAFit1 can partly prevent glucose uptake impairment by glucocorticoids, suggesting that FKBP51 might be a key factor in glucocorticoid-induced IR.
Collapse
Affiliation(s)
- Cherno O Sidibeh
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Maria J Pereira
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Xesus M Abalo
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Gretha J Boersma
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Stanko Skrtic
- AstraZeneca R&D, Mölndal, Sweden
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Per Lundkvist
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Felix Hausch
- Institute of Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Germany
| | | | - Jan W Eriksson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
17
|
Bootman MD, Allman S, Rietdorf K, Bultynck G. Deleterious effects of calcium indicators within cells; an inconvenient truth. Cell Calcium 2018; 73:82-87. [DOI: 10.1016/j.ceca.2018.04.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 04/03/2018] [Accepted: 04/11/2018] [Indexed: 01/20/2023]
|
18
|
Jaldin-Fincati JR, Pavarotti M, Frendo-Cumbo S, Bilan PJ, Klip A. Update on GLUT4 Vesicle Traffic: A Cornerstone of Insulin Action. Trends Endocrinol Metab 2017; 28:597-611. [PMID: 28602209 DOI: 10.1016/j.tem.2017.05.002] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/08/2017] [Accepted: 05/09/2017] [Indexed: 12/20/2022]
Abstract
Glucose transport is rate limiting for dietary glucose utilization by muscle and fat. The glucose transporter GLUT4 is dynamically sorted and retained intracellularly and redistributes to the plasma membrane (PM) by insulin-regulated vesicular traffic, or 'GLUT4 translocation'. Here we emphasize recent findings in GLUT4 translocation research. The application of total internal reflection fluorescence microscopy (TIRFM) has increased our understanding of insulin-regulated events beneath the PM, such as vesicle tethering and membrane fusion. We describe recent findings on Akt-targeted Rab GTPase-activating proteins (GAPs) (TBC1D1, TBC1D4, TBC1D13) and downstream Rab GTPases (Rab8a, Rab10, Rab13, Rab14, and their effectors) along with the input of Rac1 and actin filaments, molecular motors [myosinVa (MyoVa), myosin1c (Myo1c), myosinIIA (MyoIIA)], and membrane fusion regulators (syntaxin4, munc18c, Doc2b). Collectively these findings reveal novel events in insulin-regulated GLUT4 traffic.
Collapse
Affiliation(s)
| | - Martin Pavarotti
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5J 2L4, Canada; IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza 5500, Argentina
| | - Scott Frendo-Cumbo
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5J 2L4, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Philip J Bilan
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5J 2L4, Canada
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5J 2L4, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
19
|
Oxidative stress and calcium dysregulation by palmitate in type 2 diabetes. Exp Mol Med 2017; 49:e291. [PMID: 28154371 PMCID: PMC5336562 DOI: 10.1038/emm.2016.157] [Citation(s) in RCA: 227] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 10/07/2016] [Accepted: 10/16/2016] [Indexed: 12/12/2022] Open
Abstract
Free fatty acids (FFAs) are important substrates for mitochondrial oxidative metabolism and ATP synthesis but also cause serious stress to various tissues, contributing to the development of metabolic diseases. CD36 is a major mediator of cellular FFA uptake. Inside the cell, saturated FFAs are able to induce the production of cytosolic and mitochondrial reactive oxygen species (ROS), which can be prevented by co-exposure to unsaturated FFAs. There are close connections between oxidative stress and organellar Ca2+ homeostasis. Highly oxidative conditions induced by palmitate trigger aberrant endoplasmic reticulum (ER) Ca2+ release and thereby deplete ER Ca2+ stores. The resulting ER Ca2+ deficiency impairs chaperones of the protein folding machinery, leading to the accumulation of misfolded proteins. This ER stress may further aggravate oxidative stress by augmenting ER ROS production. Secondary to ER Ca2+ release, cytosolic and mitochondrial matrix Ca2+ concentrations can also be altered. In addition, plasmalemmal ion channels operated by ER Ca2+ depletion mediate persistent Ca2+ influx, further impairing cytosolic and mitochondrial Ca2+ homeostasis. Mitochondrial Ca2+ overload causes superoxide production and functional impairment, culminating in apoptosis. This vicious cycle of lipotoxicity occurs in multiple tissues, resulting in β-cell failure and insulin resistance in target tissues, and further aggravates diabetic complications.
Collapse
|
20
|
Kawabata K, Kitamura K, Irie K, Naruse S, Matsuura T, Uemae T, Taira S, Ohigashi H, Murakami S, Takahashi M, Kaido Y, Kawakami B. Triterpenoids Isolated from Ziziphus jujuba Enhance Glucose Uptake Activity in Skeletal Muscle Cells. J Nutr Sci Vitaminol (Tokyo) 2017; 63:193-199. [PMID: 28757534 DOI: 10.3177/jnsv.63.193] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Jujube (Ziziphus jujuba Mill.), a traditional folk medicine and functional food in China and South Korea, is known for its beneficial properties, which include anti-cancer, anti-oxidative, and anti-obesity effects. To assess the anti-hyperglycemic effect of jujube in this study, we investigated the glucose uptake-promoting activity of jujube in rat L6 myotubes. After determining that the jujube extract induces muscle glucose uptake, we identified the following active compounds by bioassay-guided fractionation: betulonic acid, betulinic acid, and oleanonic acid. Ursonic acid, known to be present in jujube, was semi-synthesized from ursolic acid and also observed to enhance glucose uptake. These four triterpenic acids induced glucose uptake in a glucose transporter 4-dependent manner. Comparison experiments of jujube fruits from three countries, namely, China, South Korea, and Japan, revealed that Japanese jujube has a higher content of active triterpenoids and is the most potent enhancer of glucose uptake.
Collapse
Affiliation(s)
- Kyuichi Kawabata
- Department of Bioscience, Faculty of Biotechnology, Fukui Prefectural University
| | - Kenji Kitamura
- Department of Bioscience, Faculty of Biotechnology, Fukui Prefectural University
| | - Kazuhiro Irie
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University
| | - Shoma Naruse
- Department of Bioscience, Faculty of Biotechnology, Fukui Prefectural University
| | - Tomohiro Matsuura
- Department of Bioscience, Faculty of Biotechnology, Fukui Prefectural University
| | - Tomoyuki Uemae
- Department of Bioscience, Faculty of Biotechnology, Fukui Prefectural University
| | - Shu Taira
- Department of Bioscience, Faculty of Biotechnology, Fukui Prefectural University
| | - Hajime Ohigashi
- Department of Bioscience, Faculty of Biotechnology, Fukui Prefectural University
| | - Shigeru Murakami
- Department of Bioscience, Faculty of Biotechnology, Fukui Prefectural University
| | - Masakazu Takahashi
- Department of Bioscience, Faculty of Biotechnology, Fukui Prefectural University
| | | | | |
Collapse
|
21
|
Divolis G, Mavroeidi P, Mavrofrydi O, Papazafiri P. Differential effects of calcium on PI3K-Akt and HIF-1α survival pathways. Cell Biol Toxicol 2016; 32:437-49. [PMID: 27344565 DOI: 10.1007/s10565-016-9345-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/20/2016] [Indexed: 12/19/2022]
Abstract
Calcium signaling participates in the regulation of numberless cellular functions including cell cycle progression and cellular migration, important processes for cancer expansion. Cancer cell growth, migration, and invasion are typically supported by PI3K/Akt activation, while a hypoxic environment is critical in cancer development. Accordingly, in the present study, we aimed at investigating whether perturbations in calcium homeostasis induce alterations of HIF-1α and activate Akt levels in epithelial A549 and A431 cells. Survival was drastically reduced in the presence of calcium chelator BAPTA-AM and thapsigargin, a SERCA inhibitor inducing store-operated calcium entry, to a lesser extent. Calcium chelation provoked a transient but strong upregulation of HIF-1α protein levels and accumulation in the nucleus, whereas in the presence of thapsigargin, HIF-1α levels were rapidly abolished before reaching and exceeding control levels. Despite cell death, calcium chelation merely inhibited Akt, which was significantly activated in the presence of thapsigargin. Moreover, when store-operated calcium entry was simulated by reintroducing calcium ions in cell suspensions, Akt was rapidly activated in the absence of any growth factor. These data further underscore the growing importance of calcium entry and directly link this elementary event of calcium homeostasis to the Akt pathway, which is commonly deregulated in cancer.
Collapse
Affiliation(s)
- Georgios Divolis
- Division of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Panepistimiopolis, 15784, Athens, Greece.,Center for Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation, Academy of Athens, Soranou Efesiou 4, 11527, Athens, Greece
| | - Panagiota Mavroeidi
- Division of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Panepistimiopolis, 15784, Athens, Greece
| | - Olga Mavrofrydi
- Division of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Panepistimiopolis, 15784, Athens, Greece
| | - Panagiota Papazafiri
- Division of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Panepistimiopolis, 15784, Athens, Greece.
| |
Collapse
|
22
|
Abstract
Aims There are reports that ataxia telangiectasia mutated (ATM) can activate the AMP-activated protein kinase (AMPK) and also Akt, two kinases that play integral parts in cardioprotection and metabolic function. We hypothesized that chloroquine and resveratrol, both known ATM activators, would also activate AMPK and Akt. Main methods Phosphorylation of AMPK and Akt was assessed after C2C12 myotubes were exposed to chloroquine or resveratrol. Additional experiments were done in cells expressing shRNA against ATM or in the presence of the ATM inhibitor KU55933. The effects of chloroquine on intracellular calcium were assessed with the fluorescent probe Calcium Green-1 AM. Key findings 0.5 mM chloroquine increased AMPK phosphorylation by nearly four-fold (P < 0.05), and 0.25 mM chloroquine roughly doubled Akt phosphorylation (P < 0.05). Chloroquine also increased autophosphorylation of ATM by ∼50% (P < 0.05). Resveratrol (0.15 mM) increased AMPK phosphorylation about three-fold (P < 0.05) but in contrast to chloroquine sharply decreased Akt phosphorylation. Chloroquine increased AMPK and Akt phosphorylation in myotubes expressing shRNA against ATM that reduced ATM protein levels by about 90%. Likewise, chloroquine-stimulated phosphorylation of AMPK and Akt and resveratrol-stimulated phosphorylation of AMPK were not altered by inhibition of ATM. Chloroquine decreased intracellular calcium by >50% concomitant with a decrease in glucose transport. Significance These ATM-independent effects of chloroquine on AMPK and Akt and the additional effect to decrease intracellular calcium are likely to partially underlie the positive metabolic effects of chloroquine that have been reported in the literature.
Collapse
|
23
|
Sánchez JC, Rivera RA, Muñoz LV. TRPV4 Channels in Human White Adipocytes: Electrophysiological Characterization and Regulation by Insulin. J Cell Physiol 2015; 231:954-63. [PMID: 26381274 DOI: 10.1002/jcp.25187] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 09/03/2015] [Indexed: 02/05/2023]
Abstract
Intracellular calcium homeostasis in adipocytes is important for the regulation of several functions and is involved in pathological changes in obesity and other associated diseases. Transient Receptor Potential Vanilloid 4 (TRPV4) channels are an important route for calcium entry that operates in a variety of cells and intervenes in a number of functions. In this study, the expression and operation of TRPV4 channels in human cultured adipocytes was evaluated using RT-PCR, Western blotting, the whole-cell patch-clamp technique and fluorescence measurements to characterize these channels and determine intracellular calcium responses. Both the hypoosmolarity and 4alpha-phorbol-didecanoate (4αPDD), a specific TRPV4 agonist, induced a similar HC-067047-sensitive current, which was predominantly inward, and an intracellular Ca(2+) concentration increase, which was exclusively dependent on extracellular calcium, and membrane depolarization. The current had a reverse potential of +31 ± 6 mV and exhibited preferential permeability to Ca(2+) . Insulin, which regulates metabolic homeostasis in adipocytes, attenuated the TRPV4-mediated effects. These results confirm the function of TRPV4 in human cultured adipocytes and its regulation by insulin. J. Cell. Physiol. 231: 954-963, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Julio C Sánchez
- Facultad Ciencias de la Salud, Universidad Tecnológica de Pereira, Pereira, Colombia
| | - Ricardo A Rivera
- Facultad Ciencias de la Salud, Universidad Tecnológica de Pereira, Pereira, Colombia
| | - Laura V Muñoz
- Facultad Ciencias de la Salud, Universidad Tecnológica de Pereira, Pereira, Colombia
| |
Collapse
|
24
|
Bridle KR, Sobbe AL, de Guzman CE, Santrampurwala N, Jaskowski LA, Clouston AD, Campbell CM, Nathan Subramaniam V, Crawford DHG. Lack of efficacy of mTOR inhibitors and ACE pathway inhibitors as antifibrotic agents in evolving and established fibrosis in Mdr2⁻/⁻ mice. Liver Int 2015; 35:1451-63. [PMID: 24517519 DOI: 10.1111/liv.12494] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 02/04/2014] [Indexed: 02/13/2023]
Abstract
BACKGROUND & AIMS Mammalian target of rapamycin and angiotensin-converting enzyme inhibition has been shown to have antifibrotic activity in models of liver fibrosis. The aim of our study was to determine the efficacy of rapamycin, everolimus, irbesartan and captopril, alone and in combination, as antifibrotic agents in the Mdr2(-/-) model of cholestasis both in early injury and established disease. METHODS Mdr2(-/-) mice were treated for 4 weeks with vehicle, rapamycin (1 mg/kg) or everolimus (5 mg/kg) every second day or with captopril (30 mg/kg/day), irbesartan (10 mg/kg/day) or vehicle. Further groups of 3-week-old Mdr2(-/-) mice were treated with rapamycin and irbesartan in combination (1 mg/kg/day and 10 mg/kg/day) or with rapamycin (2 mg/kg/day) for 4 weeks. Liver injury and fibrosis were compared between treated and untreated animals. RESULTS There were no significant improvements in liver injury, histology, hepatic hydroxyproline or profibrogenic gene expression following treatment with rapamycin, everolimus, captopril or irbesartan at any time point studied. Likewise, there were no improvements in liver histology or profibrogenic gene expression following combination therapy or high-dose rapamycin treatment. CONCLUSIONS The antifibrotic effects of rapamycin, everolimus, captopril and irbesartan seen in other models of fibrosis were not replicated in the Mdr2(-/-) model in this study. This highlights the clear need to test specific antifibrotic agents in a number of different animal models. We believe this animal model is ideal to study usefulness of antifibrotic agents in cholestatic liver disease because of the similarity in genetics and hepatic histopathology to human cholestatic liver disease.
Collapse
Affiliation(s)
- Kim R Bridle
- The University of Queensland School of Medicine and the Gallipoli Medical Research Foundation, Greenslopes Private Hospital, Envoi Specialist Pathologists and The Queensland Institute of Medical Research, Brisbane, Qld, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Gutiérrez T, Parra V, Troncoso R, Pennanen C, Contreras-Ferrat A, Vasquez-Trincado C, Morales PE, Lopez-Crisosto C, Sotomayor-Flores C, Chiong M, Rothermel BA, Lavandero S. Alteration in mitochondrial Ca(2+) uptake disrupts insulin signaling in hypertrophic cardiomyocytes. Cell Commun Signal 2014; 12:68. [PMID: 25376904 PMCID: PMC4234850 DOI: 10.1186/s12964-014-0068-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 10/14/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cardiac hypertrophy is characterized by alterations in both cardiac bioenergetics and insulin sensitivity. Insulin promotes glucose uptake by cardiomyocytes and its use as a substrate for glycolysis and mitochondrial oxidation in order to maintain the high cardiac energy demands. Insulin stimulates Ca(2+) release from the endoplasmic reticulum, however, how this translates to changes in mitochondrial metabolism in either healthy or hypertrophic cardiomyocytes is not fully understood. RESULTS In the present study we investigated insulin-dependent mitochondrial Ca(2+) signaling in normal and norepinephrine or insulin like growth factor-1-induced hypertrophic cardiomyocytes. Using mitochondrion-selective Ca(2+)-fluorescent probes we showed that insulin increases mitochondrial Ca(2+) levels. This signal was inhibited by the pharmacological blockade of either the inositol 1,4,5-triphosphate receptor or the mitochondrial Ca(2+) uniporter, as well as by siRNA-dependent mitochondrial Ca(2+) uniporter knockdown. Norepinephrine-stimulated cardiomyocytes showed a significant decrease in endoplasmic reticulum-mitochondrial contacts compared to either control or insulin like growth factor-1-stimulated cells. This resulted in a reduction in mitochondrial Ca(2+) uptake, Akt activation, glucose uptake and oxygen consumption in response to insulin. Blocking mitochondrial Ca(2+) uptake was sufficient to mimic the effect of norepinephrine-induced cardiomyocyte hypertrophy on insulin signaling. CONCLUSIONS Mitochondrial Ca(2+) uptake is a key event in insulin signaling and metabolism in cardiomyocytes.
Collapse
Affiliation(s)
- Tomás Gutiérrez
- Advanced Center for Chronic Disease (ACCDiS), Facultad de Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago, 838049, Chile.
| | - Valentina Parra
- Advanced Center for Chronic Disease (ACCDiS), Facultad de Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago, 838049, Chile.
- Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, TX, 75390-8573, USA.
| | - Rodrigo Troncoso
- Advanced Center for Chronic Disease (ACCDiS), Facultad de Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago, 838049, Chile.
- Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, 7830490, Chile.
| | - Christian Pennanen
- Advanced Center for Chronic Disease (ACCDiS), Facultad de Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago, 838049, Chile.
| | - Ariel Contreras-Ferrat
- Advanced Center for Chronic Disease (ACCDiS), Facultad de Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago, 838049, Chile.
- Institute for Research in Dental Science, Faculty of Dentistry, Universidad de Chile, Santiago, 838049, Chile.
| | - César Vasquez-Trincado
- Advanced Center for Chronic Disease (ACCDiS), Facultad de Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago, 838049, Chile.
| | - Pablo E Morales
- Advanced Center for Chronic Disease (ACCDiS), Facultad de Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago, 838049, Chile.
| | - Camila Lopez-Crisosto
- Advanced Center for Chronic Disease (ACCDiS), Facultad de Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago, 838049, Chile.
| | - Cristian Sotomayor-Flores
- Advanced Center for Chronic Disease (ACCDiS), Facultad de Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago, 838049, Chile.
| | - Mario Chiong
- Advanced Center for Chronic Disease (ACCDiS), Facultad de Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago, 838049, Chile.
- Centro de Estudios Moleculares de la Célula, Facultad de Medicina, Universidad de Chile, Santiago, 838049, Chile.
| | - Beverly A Rothermel
- Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, TX, 75390-8573, USA.
| | - Sergio Lavandero
- Advanced Center for Chronic Disease (ACCDiS), Facultad de Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago, 838049, Chile.
- Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, TX, 75390-8573, USA.
- Centro de Estudios Moleculares de la Célula, Facultad de Medicina, Universidad de Chile, Santiago, 838049, Chile.
| |
Collapse
|
26
|
Galione A. A primer of NAADP-mediated Ca(2+) signalling: From sea urchin eggs to mammalian cells. Cell Calcium 2014; 58:27-47. [PMID: 25449298 DOI: 10.1016/j.ceca.2014.09.010] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 09/28/2014] [Accepted: 09/29/2014] [Indexed: 02/04/2023]
Abstract
Since the discovery of the Ca(2+) mobilizing effects of the pyridine nucleotide metabolite, nicotinic acid adenine dinucleotide phosphate (NAADP), this molecule has been demonstrated to function as a Ca(2+) mobilizing intracellular messenger in a wide range of cell types. In this review, I will briefly summarize the distinct principles behind NAADP-mediated Ca(2+) signalling before going on to outline the role of this messenger in the physiology of specific cell types. Central to the discussion here is the finding that NAADP principally mobilizes Ca(2+) from acidic organelles such as lysosomes and it is this property that allows NAADP to play a unique role in intracellular Ca(2+) signalling. Lysosomes and related organelles are small Ca(2+) stores but importantly may also initiate a two-way dialogue with other Ca(2+) storage organelles to amplify Ca(2+) release, and may be strategically localized to influence localized Ca(2+) signalling microdomains. The study of NAADP signalling has created a new and fruitful focus on the lysosome and endolysosomal system as major players in calcium signalling and pathophysiology.
Collapse
Affiliation(s)
- Antony Galione
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.
| |
Collapse
|
27
|
Li J, Cantley J, Burchfield JG, Meoli CC, Stöckli J, Whitworth PT, Pant H, Chaudhuri R, Groffen AJA, Verhage M, James DE. DOC2 isoforms play dual roles in insulin secretion and insulin-stimulated glucose uptake. Diabetologia 2014; 57:2173-82. [PMID: 25005332 DOI: 10.1007/s00125-014-3312-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 05/28/2014] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS Glucose-stimulated insulin secretion (GSIS) and insulin-stimulated glucose uptake are processes that rely on regulated intracellular vesicle transport and vesicle fusion with the plasma membrane. DOC2A and DOC2B are calcium-sensitive proteins that were identified as key components of vesicle exocytosis in neurons. Our aim was to investigate the role of DOC2 isoforms in glucose homeostasis, insulin secretion and insulin action. METHODS DOC2 expression was measured by RT-PCR and western blotting. Body weight, glucose tolerance, insulin action and GSIS were assessed in wild-type (WT), Doc2a (-/-) (Doc2aKO), Doc2b (-/-) (Doc2bKO) and Doc2a (-/-)/Doc2b (-/-) (Doc2a/Doc2bKO) mice in vivo. In vitro GSIS and glucose uptake were assessed in isolated tissues, and exocytotic proteins measured by western blotting. GLUT4 translocation was assessed by epifluorescence microscopy. RESULTS Doc2b mRNA was detected in all tissues tested, whereas Doc2a was only detected in islets and the brain. Doc2aKO and Doc2bKO mice had minor glucose intolerance, while Doc2a/Doc2bKO mice showed pronounced glucose intolerance. GSIS was markedly impaired in Doc2a/Doc2bKO mice in vivo, and in isolated Doc2a/Doc2bKO islets in vitro. In contrast, Doc2bKO mice had only subtle defects in insulin secretion in vivo. Insulin action was impaired to a similar degree in both Doc2bKO and Doc2a/Doc2bKO mice. In vitro insulin-stimulated glucose transport and GLUT4 vesicle fusion were defective in adipocytes derived from Doc2bKO mice. Surprisingly, insulin action was not altered in muscle isolated from DOC2-null mice. CONCLUSIONS/INTERPRETATION Our study identifies a critical role for DOC2B in insulin-stimulated glucose uptake in adipocytes, and for the synergistic regulation of GSIS by DOC2A and DOC2B in beta cells.
Collapse
Affiliation(s)
- Jia Li
- Diabetes and Obesity Research Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Contreras-Ferrat A, Lavandero S, Jaimovich E, Klip A. Calcium signaling in insulin action on striated muscle. Cell Calcium 2014; 56:390-6. [PMID: 25224502 DOI: 10.1016/j.ceca.2014.08.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 08/24/2014] [Accepted: 08/26/2014] [Indexed: 02/07/2023]
Abstract
Striated muscles (skeletal and cardiac) are major physiological targets of insulin and this hormone triggers complex signaling pathways regulating cell growth and energy metabolism. Insulin increases glucose uptake into muscle cells by stimulating glucose transporter (GLUT4) translocation from intracellular compartments to the cell surface. The canonical insulin-triggered signaling cascade controlling this process is constituted by well-mapped tyrosine, lipid and serine/threonine phosphorylation reactions. In parallel to these signals, recent findings reveal insulin-dependent Ca(2+) mobilization in skeletal muscle cells and cardiomyocytes. Specifically, insulin activates the sarco-endoplasmic reticulum (SER) channels that release Ca(2+) into the cytosol i.e., the Ryanodine Receptor (RyR) and the inositol 1,4,5-triphosphate receptor (IP3R). In skeletal muscle cells, a rapid, insulin-triggered Ca(2+) release occurs through RyR, that is brought about upon S-glutathionylation of cysteine residues in the channel by reactive oxygen species (ROS) produced by the early activation of the NADPH oxidase (NOX2). In cardiomyocytes insulin induces a fast and transient increase in cytoplasmic [Ca(2+)]i trough L-type Ca(2+) channels activation. In both cell types, a relatively slower Ca(2+) release also occurs through IP3R activation, and is required for GLUT4 translocation and glucose uptake. The insulin-dependent Ca(2+) released from IP3R of skeletal muscle also promotes mitochondrial Ca(2+) uptake. We review here these actions of insulin on intracellular Ca(2+) channel activation and their impact on GLUT4 traffic in muscle cells, as well as other implications of insulin-dependent Ca(2+) release from the SER.
Collapse
Affiliation(s)
- A Contreras-Ferrat
- Center for Molecular Studies of the Cell (CEMC), Faculty of Medicine, Chile; Advanced Center for Chronic Disease (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Chile; Institute for Research in Dental Sciences, Faculty of Dentistry, University of Chile, Santiago 8380492, Chile.
| | - S Lavandero
- Center for Molecular Studies of the Cell (CEMC), Faculty of Medicine, Chile; Advanced Center for Chronic Disease (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Chile
| | - E Jaimovich
- Center for Molecular Studies of the Cell (CEMC), Faculty of Medicine, Chile
| | - A Klip
- The Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
| |
Collapse
|
29
|
Abstract
Insulin stimulates leptin secretion through the PI3K/Akt, but not the MAPK, pathway. Although Ca2+ alone does not trigger leptin secretion, it is required for robust Akt phosphorylation and leptin secretion.
Collapse
|
30
|
Contreras-Ferrat A, Llanos P, Vásquez C, Espinosa A, Osorio-Fuentealba C, Arias-Calderon M, Lavandero S, Klip A, Hidalgo C, Jaimovich E. Insulin elicits a ROS-activated and an IP₃-dependent Ca²⁺ release, which both impinge on GLUT4 translocation. J Cell Sci 2014; 127:1911-23. [PMID: 24569874 DOI: 10.1242/jcs.138982] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Insulin signaling includes generation of low levels of H2O2; however, its origin and contribution to insulin-stimulated glucose transport are unknown. We tested the impact of H2O2 on insulin-dependent glucose transport and GLUT4 translocation in skeletal muscle cells. H2O2 increased the translocation of GLUT4 with an exofacial Myc-epitope tag between the first and second transmembrane domains (GLUT4myc), an effect additive to that of insulin. The anti-oxidants N-acetyl L-cysteine and Trolox, the p47(phox)-NOX2 NADPH oxidase inhibitory peptide gp91-ds-tat or p47(phox) knockdown each reduced insulin-dependent GLUT4myc translocation. Importantly, gp91-ds-tat suppressed insulin-dependent H2O2 production. A ryanodine receptor (RyR) channel agonist stimulated GLUT4myc translocation and insulin stimulated RyR1-mediated Ca(2+) release by promoting RyR1 S-glutathionylation. This pathway acts in parallel to insulin-mediated stimulation of inositol-1,4,5-trisphosphate (IP3)-activated Ca(2+) channels, in response to activation of phosphatidylinositol 3-kinase and its downstream target phospholipase C, resulting in Ca(2+) transfer to the mitochondria. An inhibitor of IP3 receptors, Xestospongin B, reduced both insulin-dependent IP3 production and GLUT4myc translocation. We propose that, in addition to the canonical α,β phosphatidylinositol 3-kinase to Akt pathway, insulin engages both RyR-mediated Ca(2+) release and IP3-receptor-mediated mitochondrial Ca(2+) uptake, and that these signals jointly stimulate glucose uptake.
Collapse
Affiliation(s)
- Ariel Contreras-Ferrat
- Centro de estudios Moleculares de la Célula, Facultad de Medicina; Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Turner N, Cooney GJ, Kraegen EW, Bruce CR. Fatty acid metabolism, energy expenditure and insulin resistance in muscle. J Endocrinol 2014; 220:T61-79. [PMID: 24323910 DOI: 10.1530/joe-13-0397] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Fatty acids (FAs) are essential elements of all cells and have significant roles as energy substrates, components of cellular structure and signalling molecules. The storage of excess energy intake as fat in adipose tissue is an evolutionary advantage aimed at protecting against starvation, but in much of today's world, humans are faced with an unlimited availability of food, and the excessive accumulation of fat is now a major risk for human health, especially the development of type 2 diabetes (T2D). Since the first recognition of the association between fat accumulation, reduced insulin action and increased risk of T2D, several mechanisms have been proposed to link excess FA availability to reduced insulin action, with some of them being competing or contradictory. This review summarises the evidence for these mechanisms in the context of excess dietary FAs generating insulin resistance in muscle, the major tissue involved in insulin-stimulated disposal of blood glucose. It also outlines potential problems with models and measurements that may hinder as well as help improve our understanding of the links between FAs and insulin action.
Collapse
Affiliation(s)
- Nigel Turner
- Department of Pharmacology School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia Diabetes and Obesity Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, New South Wales 2010, Australia St Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia Department of Physiology, Monash University, Clayton, Victoria 3800, Australia
| | | | | | | |
Collapse
|
32
|
Stall R, Ramos J, Kent Fulcher F, Patel YM. Regulation of myosin IIA and filamentous actin during insulin-stimulated glucose uptake in 3T3-L1 adipocytes. Exp Cell Res 2013; 322:81-8. [PMID: 24374234 DOI: 10.1016/j.yexcr.2013.12.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 12/04/2013] [Accepted: 12/08/2013] [Indexed: 11/18/2022]
Abstract
Insulin stimulated glucose uptake requires the colocalization of myosin IIA (MyoIIA) and the insulin-responsive glucose transporter 4 (GLUT4) at the plasma membrane for proper GLUT4 fusion. MyoIIA facilitates filamentous actin (F-actin) reorganization in various cell types. In adipocytes F-actin reorganization is required for insulin-stimulated glucose uptake. What is not known is whether MyoIIA interacts with F-actin to regulate insulin-induced GLUT4 fusion at the plasma membrane. To elucidate the relationship between MyoIIA and F-actin, we examined the colocalization of MyoIIA and F-actin at the plasma membrane upon insulin stimulation as well as the regulation of this interaction. Our findings demonstrated that MyoIIA and F-actin colocalized at the site of GLUT4 fusion with the plasma membrane upon insulin stimulation. Furthermore, inhibition of MyoII with blebbistatin impaired F-actin localization at the plasma membrane. Next we examined the regulatory role of calcium in MyoIIA-F-actin colocalization. Reduced calcium or calmodulin levels decreased colocalization of MyoIIA and F-actin at the plasma membrane. While calcium alone can translocate MyoIIA it did not stimulate F-actin accumulation at the plasma membrane. Taken together, we established that while MyoIIA activity is required for F-actin localization at the plasma membrane, it alone is insufficient to localize F-actin to the plasma membrane.
Collapse
Affiliation(s)
- Richard Stall
- Department of Biology, University of North Carolina at Greensboro, 312 Eberhart Building, Greensboro, NC 27412, USA
| | - Joseph Ramos
- Department of Biology, University of North Carolina at Greensboro, 312 Eberhart Building, Greensboro, NC 27412, USA
| | - F Kent Fulcher
- Department of Biology, University of North Carolina at Greensboro, 312 Eberhart Building, Greensboro, NC 27412, USA
| | - Yashomati M Patel
- Department of Biology, University of North Carolina at Greensboro, 312 Eberhart Building, Greensboro, NC 27412, USA.
| |
Collapse
|
33
|
Woody S, Stall R, Ramos J, Patel YM. Regulation of myosin light chain kinase during insulin-stimulated glucose uptake in 3T3-L1 adipocytes. PLoS One 2013; 8:e77248. [PMID: 24116218 PMCID: PMC3792908 DOI: 10.1371/journal.pone.0077248] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 09/09/2013] [Indexed: 11/25/2022] Open
Abstract
Myosin II (MyoII) is required for insulin-responsive glucose transporter 4 (GLUT4)-mediated glucose uptake in 3T3-L1 adipocytes. Our previous studies have shown that insulin signaling stimulates phosphorylation of the regulatory light chain (RLC) of MyoIIA via myosin light chain kinase (MLCK). The experiments described here delineate upstream regulators of MLCK during insulin-stimulated glucose uptake. Since 3T3-L1 adipocytes express two MyoII isoforms, we wanted to determine which isoform was required for insulin-stimulated glucose uptake. Using a siRNA approach, we demonstrate that a 60% decrease in MyoIIA protein expression resulted in a 40% inhibition of insulin-stimulated glucose uptake. We also show that insulin signaling stimulates the phosphorylation of MLCK. We further show that MLCK can be activated by calcium as well as signaling pathways. We demonstrate that adipocytes treated with the calcium chelating agent, 1,2-b (iso-aminophenoxy) ethane-N,N,N',N'-tetra acetic acid, (BAPTA) (in the presence of insulin) impaired the insulin-induced phosphorylation of MLCK by 52% and the RLC of MyoIIA by 45% as well as impairing the recruitment of MyoIIA to the plasma membrane when compared to cells treated with insulin alone. We further show that the calcium ionophore, A23187 alone stimulated the phosphorylation of MLCK and the RLC associated with MyoIIA to the same extent as insulin. To identify signaling pathways that might regulate MLCK, we examined ERK and CaMKII. Inhibition of ERK2 impaired phosphorylation of MLCK and insulin-stimulated glucose uptake. In contrast, while inhibition of CaMKII did inhibit phosphorylation of the RLC associated with MyoIIA, inhibition of CAMKIIδ did not impair MLCK phosphorylation or translocation to the plasma membrane or glucose uptake. Collectively, our results are the first to delineate a role for calcium and ERK in the activation of MLCK and thus MyoIIA during insulin-stimulated glucose uptake in 3T3-L1 adipocytes.
Collapse
Affiliation(s)
- Shelly Woody
- Department of Biology, University of North Carolina at Greensboro, Greensboro, North Carolina, United States of America
| | - Richard Stall
- Department of Biology, University of North Carolina at Greensboro, Greensboro, North Carolina, United States of America
| | - Joseph Ramos
- Department of Biology, University of North Carolina at Greensboro, Greensboro, North Carolina, United States of America
| | - Yashomati M. Patel
- Department of Biology, University of North Carolina at Greensboro, Greensboro, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
34
|
Chen KE, Richards AA, Caradoc-Davies TT, Vajjhala PR, Robin G, Lua LHL, Hill JM, Schroder K, Sweet MJ, Kellie S, Kobe B, Martin J. The structure of the caspase recruitment domain of BinCARD reveals that all three cysteines can be oxidized. ACTA CRYSTALLOGRAPHICA SECTION D: BIOLOGICAL CRYSTALLOGRAPHY 2013; 69:774-84. [PMID: 23633586 DOI: 10.1107/s0907444913001558] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 01/16/2013] [Indexed: 11/10/2022]
Abstract
The caspase recruitment domain (CARD) is present in death-domain superfamily proteins involved in inflammation and apoptosis. BinCARD is named for its ability to interact with Bcl10 and inhibit downstream signalling. Human BinCARD is expressed as two isoforms that encode the same N-terminal CARD region but which differ considerably in their C-termini. Both isoforms are expressed in immune cells, although BinCARD-2 is much more highly expressed. Crystals of the CARD fold common to both had low symmetry (space group P1). Molecular replacement was unsuccessful in this low-symmetry space group and, as the construct contains no methionines, first one and then two residues were engineered to methionine for MAD phasing. The double-methionine variant was produced as a selenomethionine derivative, which was crystallized and the structure was solved using data measured at two wavelengths. The crystal structures of the native and selenomethionine double mutant were refined to high resolution (1.58 and 1.40 Å resolution, respectively), revealing the presence of a cis-peptide bond between Tyr39 and Pro40. Unexpectedly, the native crystal structure revealed that all three cysteines were oxidized. The mitochondrial localization of BinCARD-2 and the susceptibility of its CARD region to redox modification points to the intriguing possibility of a redox-regulatory role.
Collapse
Affiliation(s)
- Kai En Chen
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland 4072, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Yu H, Rathore SS, Davis EM, Ouyang Y, Shen J. Doc2b promotes GLUT4 exocytosis by activating the SNARE-mediated fusion reaction in a calcium- and membrane bending-dependent manner. Mol Biol Cell 2013; 24:1176-84. [PMID: 23427263 PMCID: PMC3623638 DOI: 10.1091/mbc.e12-11-0810] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Reconstitution of GLUT4 vesicle fusion in a defined fusion system shows that the C2-domain factor Doc2b activates the SNARE-dependent fusion reaction by a calcium- and membrane bending–dependent mechanism. Of importance, certain features of Doc2b function appear to be distinct from how synaptotagmin-1 promotes synaptic release. The glucose transporter GLUT4 plays a central role in maintaining body glucose homeostasis. On insulin stimulation, GLUT4-containing vesicles fuse with the plasma membrane, relocating GLUT4 from intracellular reservoirs to the cell surface to uptake excess blood glucose. The GLUT4 vesicle fusion reaction requires soluble N-ethylmaleimide–sensitive factor attachment protein receptors (SNAREs) as the core fusion engine and a group of regulatory proteins. In particular, the soluble C2-domain factor Doc2b plays a key role in GLUT4 vesicle fusion, but its molecular mechanism has been unclear. Here we reconstituted the SNARE-dependent GLUT4 vesicle fusion in a defined proteoliposome fusion system. We observed that Doc2b binds to GLUT4 exocytic SNAREs and potently accelerates the fusion kinetics in the presence of Ca2+. The stimulatory activity of Doc2b requires intact Ca2+-binding sites on both the C2A and C2B domains. Using electron microscopy, we observed that Doc2b strongly bends the membrane bilayer, and this membrane-bending activity is essential to the stimulatory function of Doc2b in fusion. These results demonstrate that Doc2b promotes GLUT4 exocytosis by accelerating the SNARE-dependent fusion reaction by a Ca2+- and membrane bending–dependent mechanism. Of importance, certain features of Doc2b function appear to be distinct from how synaptotagmin-1 promotes synaptic neurotransmitter release, suggesting that exocytic Ca2+ sensors may possess divergent mechanisms in regulating vesicle fusion.
Collapse
Affiliation(s)
- Haijia Yu
- Department of Molecular, Cellular and Developmental Biology, University of Colorado at Boulder, Boulder, CO 80309, USA
| | | | | | | | | |
Collapse
|
36
|
Chen SJ, Zhang W, Tong Q, Conrad K, Hirschler-Laszkiewicz I, Bayerl M, Kim JK, Cheung JY, Miller BA. Role of TRPM2 in cell proliferation and susceptibility to oxidative stress. Am J Physiol Cell Physiol 2013; 304:C548-60. [PMID: 23302782 DOI: 10.1152/ajpcell.00069.2012] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The transient receptor potential (TRP) channel TRPM2 is an ion channel that modulates cell survival. We report here that full-length (TRPM2-L) and short (TRPM2-S) isoform expression was significantly increased in human neuroblastoma compared with adrenal gland. To differentiate the roles of TRPM2-L and TRPM2-S in cell proliferation and survival, we established neuroblastoma SH-SY5Y cell lines stably expressing either TRPM2 isoform or empty vector. Cells expressing TRPM2-S showed significantly enhanced proliferation, downregulation of phosphatase and tensin homolog (PTEN), and increased protein kinase B (Akt) phosphorylation and cell surface glucose transporter 1 (Glut1) compared with cells expressing TRPM2-L or empty vector. ERK phosphorylation was increased, and forkhead box O 3a (FOXO3a) levels were decreased. Inhibitor studies demonstrated that enhanced proliferation was dependent on phosphatidylinositol 3-kinase/Akt, ERK, and NADPH oxidase activation. On the other hand, TRPM2-S-expressing cells were significantly more susceptible to cell death induced by low H2O2 concentrations (50-100 μM), whereas TRPM2-L-expressing cells were protected. This was associated with a significant increase in FOXO3a, MnSOD (SOD2), and membrane Glut1 in TRPM2-L-expressing cells compared with TRPM2-S expressing cells. We conclude that TRPM2 channels occupy a key role in cell proliferation and survival following oxidative stress in neuroblastoma. Our results suggest that overexpression of TRPM2-S results in increased proliferation through phosphatidylinositol 3-kinase/Akt and ERK pathways, while overexpression of TRPM2-L confers protection against oxidative stress-induced cell death through FOXO3a and SOD. TRPM2 channels may represent a novel future therapeutic target in diseases involving oxidative stress.
Collapse
Affiliation(s)
- Shu-jen Chen
- Department of Pediatrics, Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, PO Box 850, Hershey, PA 17033, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Yao Y, Suraokar M, Darnay BG, Hollier BG, Shaiken TE, Asano T, Chen CH, Chang BHJ, Lu Y, Mills GB, Sarbassov D, Mani SA, Abbruzzese JL, Reddy SAG. BSTA promotes mTORC2-mediated phosphorylation of Akt1 to suppress expression of FoxC2 and stimulate adipocyte differentiation. Sci Signal 2013; 6:ra2. [PMID: 23300339 DOI: 10.1126/scisignal.2003295] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Phosphorylation and activation of Akt1 is a crucial signaling event that promotes adipogenesis. However, neither the complex multistep process that leads to activation of Akt1 through phosphorylation at Thr³⁰⁸ and Ser⁴⁷³ nor the mechanism by which Akt1 stimulates adipogenesis is fully understood. We found that the BSD domain-containing signal transducer and Akt interactor (BSTA) promoted phosphorylation of Akt1 at Ser⁴⁷³ in various human and murine cells, and we uncovered a function for the BSD domain in BSTA-Akt1 complex formation. The mammalian target of rapamycin complex 2 (mTORC2) facilitated the phosphorylation of BSTA and its association with Akt1, and the BSTA-Akt1 interaction promoted the association of mTORC2 with Akt1 and phosphorylation of Akt1 at Ser⁴⁷³ in response to growth factor stimulation. Furthermore, analyses of bsta gene-trap murine embryonic stem cells revealed an essential function for BSTA and phosphorylation of Akt1 at Ser⁴⁷³ in promoting adipocyte differentiation, which required suppression of the expression of the gene encoding the transcription factor FoxC2. These findings indicate that BSTA is a molecular switch that promotes phosphorylation of Akt1 at Ser⁴⁷³ and reveal an mTORC2-BSTA-Akt1-FoxC2-mediated signaling mechanism that is critical for adipocyte differentiation.
Collapse
Affiliation(s)
- Yixin Yao
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Song EK, Lee YR, Kim YR, Yeom JH, Yoo CH, Kim HK, Park HM, Kang HS, Kim JS, Kim UH, Han MK. NAADP mediates insulin-stimulated glucose uptake and insulin sensitization by PPARγ in adipocytes. Cell Rep 2012. [PMID: 23177620 DOI: 10.1016/j.celrep.2012.10.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Insulin stimulates glucose uptake through the membrane translocation of GLUT4 and GLUT1. Peroxisome proliferator-activated receptor γ (PPARγ) enhances insulin sensitivity. Here, we demonstrate that insulin stimulates GLUT4 and GLUT1 translocation, and glucose uptake, by activating the signaling pathway involving nicotinic acid adenine dinucleotide phosphate (NAADP), a calcium mobilizer, in adipocytes. We also demonstrate that PPARγ mediates insulin sensitization by enhancing NAADP production through upregulation of CD38, the only enzyme identified for NAADP synthesis. Insulin produced NAADP by both CD38-dependent and -independent pathways, whereas PPARγ produced NAADP by CD38-dependent pathway. Blocking the NAADP signaling pathway abrogated both insulin-stimulated and PPARγ-induced GLUT4 and GLUT1 translocation, thereby inhibiting glucose uptake. CD38 knockout partially inhibited insulin-stimulated glucose uptake. However, CD38 knockout completely blocked PPARγ-induced glucose uptake in adipocytes and PPARγ-mediated amelioration of glucose tolerance in diabetic mice. These results demonstrated that the NAADP signaling pathway is a critical molecular target for PPARγ-mediated insulin sensitization.
Collapse
Affiliation(s)
- Eun-Kyung Song
- Department of Microbiology, Chonbuk National University Medical School, Jeonju 561-756, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Zhang JQ, Wu PF, Long LH, Chen Y, Hu ZL, Ni L, Wang F, Chen JG. Resveratrol promotes cellular glucose utilization in primary cultured cortical neurons via calcium-dependent signaling pathway. J Nutr Biochem 2012; 24:629-37. [PMID: 22819552 DOI: 10.1016/j.jnutbio.2012.02.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 02/19/2012] [Accepted: 02/28/2012] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND PURPOSE Impairment of glucose utilization contributes to neuronal degeneration of Alzheimer's disease patients. Cellular glucose utilization can be regulated by calcium-dependent signaling pathways. Resveratrol (RSV) is a plant-derived polyphenol with multiple beneficial effects, including neuroprotection and metabolic improvement. Here, we investigated the effect of RSV on neuronal calcium signal and glucose utilization. EXPERIMENTAL METHODS Primary culture of cortical neurons, calcium imaging, 2-NBDG assay and western blotting were employed to investigate RSV-mediated effects on neuronal calcium signal and glucose utilization. RESULTS RSV elevated intracellular calcium in cortical neurons via modulation of secondary messenger system including nitrous oxide, cGMP and cAMP. Secondarily, a calcium-dependent enhancement of neuronal glucose utilization after RSV treatment was observed. The effects on neuronal glucose utilization are largely dependent on RSV-induced calcium-dependent AMP-activated protein kinase activation. CONCLUSION Our findings show that activation of calcium-dependent signaling pathways by RSV may convey improvements of neuronal glucose utilization.
Collapse
Affiliation(s)
- Jun-Qi Zhang
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | | | | | | | | | | | | | | |
Collapse
|
40
|
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
|
41
|
Boller S, Joblin BA, Xu L, Item F, Trüb T, Boschetti N, Spinas GA, Niessen M. From signal transduction to signal interpretation: an alternative model for the molecular function of insulin receptor substrates. Arch Physiol Biochem 2012; 118:148-55. [PMID: 22515179 DOI: 10.3109/13813455.2012.671333] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The insulin receptor (IR) recruits adaptor proteins, so-called insulin receptor substrates (IRS), to connect with downstream signalling pathways. A family of IRS proteins was defined based on three major common structural elements: Amino-terminal PH and PTB domains that mediate protein-lipid or protein-protein interactions, mostly carboxy-terminal multiple tyrosine residues that serve as binding sites for proteins that contain one or more SH2 domains and serine/threonine-rich regions which may be recognized by negative regulators of insulin action. The current model for the role of IRS proteins therefore combines an adaptor function with the integration of mostly negative input from other signal transduction cascades allowing for modulation of signalling amplitude. In this review we propose an extended version of the adaptor model that can explain how signalling specificity could be implemented at the level of IRS proteins.
Collapse
Affiliation(s)
- Simone Boller
- Endocrinology, Diabetes and Clinical Nutrition, University Hospital of Zurich, 8091 Zurich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Tan SX, Ng Y, Meoli CC, Kumar A, Khoo PS, Fazakerley DJ, Junutula JR, Vali S, James DE, Stöckli J. Amplification and demultiplexing in insulin-regulated Akt protein kinase pathway in adipocytes. J Biol Chem 2011; 287:6128-38. [PMID: 22207758 PMCID: PMC3307283 DOI: 10.1074/jbc.m111.318238] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Akt plays a major role in insulin regulation of metabolism in muscle, fat, and liver. Here, we show that in 3T3-L1 adipocytes, Akt operates optimally over a limited dynamic range. This indicates that Akt is a highly sensitive amplification step in the pathway. With robust insulin stimulation, substantial changes in Akt phosphorylation using either pharmacologic or genetic manipulations had relatively little effect on Akt activity. By integrating these data we observed that half-maximal Akt activity was achieved at a threshold level of Akt phosphorylation corresponding to 5–22% of its full dynamic range. This behavior was also associated with lack of concordance or demultiplexing in the behavior of downstream components. Most notably, FoxO1 phosphorylation was more sensitive to insulin and did not exhibit a change in its rate of phosphorylation between 1 and 100 nm insulin compared with other substrates (AS160, TSC2, GSK3). Similar differences were observed between various insulin-regulated pathways such as GLUT4 translocation and protein synthesis. These data indicate that Akt itself is a major amplification switch in the insulin signaling pathway and that features of the pathway enable the insulin signal to be split or demultiplexed into discrete outputs. This has important implications for the role of this pathway in disease.
Collapse
Affiliation(s)
- Shi-Xiong Tan
- Diabetes and Obesity Research Program, The Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, New South Wales 2010, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ogawa A, Firth AL, Smith KA, Maliakal MV, Yuan JXJ. PDGF enhances store-operated Ca2+ entry by upregulating STIM1/Orai1 via activation of Akt/mTOR in human pulmonary arterial smooth muscle cells. Am J Physiol Cell Physiol 2011; 302:C405-11. [PMID: 22031597 DOI: 10.1152/ajpcell.00337.2011] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Platelet-derived growth factor (PDGF) and its receptor are known to be substantially elevated in lung tissues and pulmonary arterial smooth muscle cells (PASMC) isolated from patients and animals with pulmonary arterial hypertension. PDGF has been shown to phosphorylate and activate Akt and mammalian target of rapamycin (mTOR) in PASMC. In this study, we investigated the role of PDGF-mediated activation of Akt signaling in the regulation of cytosolic Ca(2+) concentration and cell proliferation. PDGF activated the Akt/mTOR pathway and, subsequently, enhanced store-operated Ca(2+) entry (SOCE) and cell proliferation in human PASMC. Inhibition of Akt attenuated the increase in cytosolic Ca(2+) concentration due to both SOCE and PASMC proliferation. This effect correlated with a significant downregulation of stromal interacting molecule (STIM) and Orai, proposed molecular correlates for SOCE in many cell types. The data from this study present a novel pathway for the regulation of Ca(2+) signaling and PASMC proliferation involving activation of Akt in response to upregulated expression of PDGF. Targeting this pathway may lead to the development of a novel therapeutic option for the treatment of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Aiko Ogawa
- Department of Clinical Science, National Hospital Organization Okayama Medical Center, Japan
| | | | | | | | | |
Collapse
|
44
|
Jung JG, Choi SE, Hwang YJ, Lee SA, Kim EK, Lee MS, Han SJ, Kim HJ, Kim DJ, Kang Y, Lee KW. Supplementation of pyruvate prevents palmitate-induced impairment of glucose uptake in C2 myotubes. Mol Cell Endocrinol 2011; 345:79-87. [PMID: 21802492 DOI: 10.1016/j.mce.2011.07.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Revised: 07/08/2011] [Accepted: 07/08/2011] [Indexed: 12/22/2022]
Abstract
Elevated fatty acid levels have been thought to contribute to insulin resistance. Repression of the glucose transporter 4 (GLUT4) gene as well as impaired GLUT4 translocation may be a mediator for fatty acid-induced insulin resistance. This study was initiated to determine whether palmitate treatment repressed GLUT4 expression, whether glucose/fatty acid metabolism influenced palmitate-induced GLUT4 gene repression (PIGR), and whether attempts to prevent PIGR restored palmitate-induced impairment of glucose uptake (PIIGU) in C2 myotubes. Not only stimulators of fatty acid oxidation, such as bezafibrate, AICAR, and TOFA, but also TCA cycle substrates, such as pyruvate, leucine/glutamine, and α-ketoisocaproate/monomethyl succinate, significantly prevented PIGR. In particular, supplementing with pyruvate through methyl pyruvate resulted in nearly complete prevention of PIIGU, whereas palmitate treatment reduced the intracellular pyruvate level. These results suggest that pyruvate depletion plays a critical role in PIGR and PIIGU; thus, pyruvate supplementation may help prevent obesity-induced insulin resistance in muscle cells.
Collapse
Affiliation(s)
- Jong Gab Jung
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Hutley LJ, Newell FS, Kim YH, Luo X, Widberg CH, Shurety W, Prins JB, Whitehead JP. A putative role for endogenous FGF-2 in FGF-1 mediated differentiation of human preadipocytes. Mol Cell Endocrinol 2011; 339:165-71. [PMID: 21539890 DOI: 10.1016/j.mce.2011.04.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2010] [Revised: 04/14/2011] [Accepted: 04/15/2011] [Indexed: 11/27/2022]
Abstract
The defining characteristic of obesity is increased adipose tissue (AT) mass following chronic positive energy supply. AT mass is determined by adipocyte number and size, which reflect proliferation and differentiation of preadipocytes and hypertrophy of pre-existing adipocytes. The molecular pathways governing AT expansion are incompletely defined. We previously reported that FGF-1 primes proliferating primary human preadipocytes (phPA), thereby increasing adipogenesis. Here we examined whether FGF-1's adipogenic actions were due to modulation of other FGFs. Treatment of phPA with FGF-1 reduced FGF-2 mRNA/protein by 80%. To examine a putative functional role we performed siRNA knockdown studies. Following FGF-2 knockdown preadipocyte proliferation was decreased and expression of adipogenic genes (PPARγ, G3PDH and adiponectin) was increased at day 1 of differentiation. These results suggest that changes in endogenous FGF-2 levels contribute to FGF-1's early adipogenic effects and highlight the complexity of the paracrine interplay between FGFs within human AT.
Collapse
Affiliation(s)
- Louise J Hutley
- Metabolic Program, Mater Medical Research Institute, Mater Health Services, South Brisbane, Queensland 4101, Australia
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Gormand A, Henriksson E, Ström K, Jensen TE, Sakamoto K, Göransson O. Regulation of AMP-activated protein kinase by LKB1 and CaMKK in adipocytes. J Cell Biochem 2011; 112:1364-75. [DOI: 10.1002/jcb.23053] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
47
|
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
|
48
|
Brandon AE, Hoy AJ, Wright LE, Turner N, Hegarty BD, Iseli TJ, Julia Xu X, Cooney GJ, Saha AK, Ruderman NB, Kraegen EW. The evolution of insulin resistance in muscle of the glucose infused rat. Arch Biochem Biophys 2011; 509:133-41. [PMID: 21420928 DOI: 10.1016/j.abb.2011.03.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2011] [Revised: 03/14/2011] [Accepted: 03/15/2011] [Indexed: 11/17/2022]
Abstract
Glucose infusion into rats causes skeletal muscle insulin resistance that initially occurs without changes in insulin signaling. The aim of the current study was to prolong glucose infusion and evaluate other events associated with the transition to muscle insulin resistance. Hyperglycemia was produced in rats by glucose infusion for 3, 5 and 8 h. The rate of infusion required to maintain hyperglycemia was reduced at 5 and 8 h. Glucose uptake into red quadriceps (RQ) and its incorporation into glycogen decreased between 3 and 5 h, further decreasing at 8 h. The earliest observed change in RQ was decreased AMPKα2 activity associated with large increases in muscle glycogen content at 3 h. Activation of the mTOR pathway occurred at 5 h. Akt phosphorylation (Ser(473)) was decreased at 8 h compared to 3 and 5, although no decrease in phosphorylation of downstream GSK-3β (Ser(9)) and AS160 (Thr(642)) was observed. White quadriceps showed a similar but delayed pattern, with insulin resistance developing by 8 h and decreased AMPKα2 activity at 5 h. These results indicate that, in the presence of a nutrient overload, alterations in muscle insulin signaling occur, but after insulin resistance develops and appropriate changes in energy/nutrient sensing pathways occur.
Collapse
Affiliation(s)
- Amanda E Brandon
- Diabetes and Obesity Program, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Grisouard J, Timper K, Radimerski TM, Frey DM, Peterli R, Kola B, Korbonits M, Herrmann P, Krähenbühl S, Zulewski H, Keller U, Müller B, Christ-Crain M. Mechanisms of metformin action on glucose transport and metabolism in human adipocytes. Biochem Pharmacol 2010; 80:1736-45. [DOI: 10.1016/j.bcp.2010.08.021] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 08/23/2010] [Accepted: 08/25/2010] [Indexed: 01/28/2023]
|
50
|
Kim SJ, Choi HJ, Jung CH, Park SS, Cho SR, Oh SJ, Kim ES. Calcium Mobilization Inhibits Lipid Accumulation During the Late Adipogenesis via Suppression of PPARγ and LXRα Signalings. Korean J Food Sci Anim Resour 2010. [DOI: 10.5851/kosfa.2010.30.5.787] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|