1
|
Sun YD, Zhang H, Li YM, Han JJ. Abnormal metabolism in hepatic stellate cells: Pandora's box of MAFLD related hepatocellular carcinoma. Biochim Biophys Acta Rev Cancer 2024; 1879:189086. [PMID: 38342420 DOI: 10.1016/j.bbcan.2024.189086] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/25/2023] [Accepted: 02/06/2024] [Indexed: 02/13/2024]
Abstract
Metabolic associated fatty liver disease (MAFLD) is a significant risk factor for the development of hepatocellular carcinoma (HCC). Hepatic stellate cells (HSCs), as key mediators in liver injury response, are believed to play a crucial role in the repair process of liver injury. However, in MAFLD patients, the normal metabolic and immunoregulatory mechanisms of HSCs become disrupted, leading to disturbances in the local microenvironment. Abnormally activated HSCs are heavily involved in the initiation and progression of HCC. The metabolic disorders and abnormal activation of HSCs not only initiate liver fibrosis but also contribute to carcinogenesis. In this review, we provide an overview of recent research progress on the relationship between the abnormal metabolism of HSCs and the local immune system in the liver, elucidating the mechanisms of immune imbalance caused by abnormally activated HSCs in MAFLD patients. Based on this understanding, we discuss the potential and challenges of metabolic-based and immunology-based mechanisms in the treatment of MAFLD-related HCC, with a specific focus on the role of HSCs in HCC progression and their potential as targets for anti-cancer therapy. This review aims to enhance researchers' understanding of the importance of HSCs in maintaining normal liver function and highlights the significance of HSCs in the progression of MAFLD-related HCC.
Collapse
Affiliation(s)
- Yuan-Dong Sun
- Department of Interventional Radiology, Shandong Cancer Hospital and Institute Affiliated Shandong First Medical University, Shandong Academy of Medical Sciences, China
| | - Hao Zhang
- Department of Interventional Radiology, Shandong Cancer Hospital and Institute Affiliated Shandong First Medical University, Shandong Academy of Medical Sciences, China
| | - Yuan-Min Li
- NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, China
| | - Jian-Jun Han
- Department of Interventional Radiology, Shandong Cancer Hospital and Institute Affiliated Shandong First Medical University, Shandong Academy of Medical Sciences, China.
| |
Collapse
|
2
|
GLUT4 translocation and dispersal operate in multiple cell types and are negatively correlated with cell size in adipocytes. Sci Rep 2022; 12:20535. [PMID: 36446811 PMCID: PMC9708847 DOI: 10.1038/s41598-022-24736-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/18/2022] [Indexed: 11/30/2022] Open
Abstract
The regulated translocation of the glucose transporter, GLUT4, to the surface of adipocytes and muscle is a key action of insulin. This is underpinned by the delivery and fusion of GLUT4-containing vesicles with the plasma membrane. Recent studies have revealed that a further action of insulin is to mediate the dispersal of GLUT4 molecules away from the site of GLUT4 vesicle fusion with the plasma membrane. Although shown in adipocytes, whether insulin-stimulated dispersal occurs in other cells and/or is exhibited by other proteins remains a matter of debate. Here we show that insulin stimulates GLUT4 dispersal in the plasma membrane of adipocytes, induced pluripotent stem cell-derived cardiomyocytes and HeLa cells, suggesting that this phenomenon is specific to GLUT4 expressed in all cell types. By contrast, insulin-stimulated dispersal of TfR was not observed in HeLa cells, suggesting that the mechanism may be unique to GLUT4. Consistent with dispersal being an important physiological mechanism, we observed that insulin-stimulated GLUT4 dispersal is reduced under conditions of insulin resistance. Adipocytes of different sizes have been shown to exhibit distinct metabolic properties: larger adipocytes exhibit reduced insulin-stimulated glucose transport compared to smaller cells. Here we show that both GLUT4 delivery to the plasma membrane and GLUT4 dispersal are reduced in larger adipocytes, supporting the hypothesis that larger adipocytes are refractory to insulin challenge compared to their smaller counterparts, even within a supposedly homogeneous population of cells.
Collapse
|
3
|
Lee HY, Lee GH, Hoang TH, Park SA, Lee J, Lim J, Sa S, Kim GE, Han JS, Kim J, Chae HJ. d-Allulose Ameliorates Hyperglycemia Through IRE1α Sulfonation-RIDD- Sirt1 Decay Axis in the Skeletal Muscle. Antioxid Redox Signal 2022; 37:229-245. [PMID: 35166127 DOI: 10.1089/ars.2021.0207] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Aims: The skeletal muscle maintains glucose disposal via insulin signaling and glucose transport. The progression of diabetes and insulin resistance is critically influenced by endoplasmic reticulum (ER) stress. d-Allulose, a low-calorie sugar substitute, has shown crucial physiological activities under conditions involving hyperglycemia and insulin resistance. However, the molecular mechanisms of d-allulose in the progression of diabetes have not been fully elucidated. Here, we evaluated the effect of d-allulose on hyperglycemia-associated ER stress responses in human skeletal myoblasts (HSkM) and db/db diabetic and high-fat diet-fed mice. Results: d-allulose effectively controlled glycemic markers such as insulin and hemoglobin A1c (HbA1c), showing anti-diabetic effects by inhibiting the disruption of insulin receptor substrate (IRS)-1 tyrosine phosphorylation and glucose transporter 4 (GLUT4) expression, in which the phosphatidylinositol-3 kinase (PI3K)/protein kinase B (Akt) pathway is involved. The levels of glucose dysmetabolism-based NADPH oxidase, such as NADPH-dependent oxidoreductase (Nox) 4, were highly increased, and their interaction with IRE1α and the resultant sulfonation-regulated IRE1-dependent decay (RIDD)-Sirt1 decay were also highly increased under diabetic conditions, which were controlled with d-allulose treatment. Skeletal muscle cells grown with a high glucose medium supplemented with d-allulose showed controlled IRE1α sulfonation-RIDD-Sirt1 decay, in which Nox4 was involved. Innovation and Conclusion: The study observations indicate that d-allulose contributes to the muscular glucose disposal in the diabetic state where ER-localized Nox4-induced IRE1α sulfonation results in the decay of Sirt1, a core factor for controlling glucose metabolism. Antioxid. Redox Signal. 37, 229-245.
Collapse
Affiliation(s)
- Hwa-Young Lee
- Department of Pharmacology and Institute of New Drug Development, Jeonbuk National University Medical School, Jeonju, South Korea.,Non-Clinical Evaluation Center Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, South Korea
| | - Geum-Hwa Lee
- Non-Clinical Evaluation Center Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, South Korea
| | - The-Hiep Hoang
- Non-Clinical Evaluation Center Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, South Korea.,Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - Seon-Ah Park
- Non-Clinical Evaluation Center Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, South Korea
| | - Juwon Lee
- School of Pharmacy, Jeonbuk National University, Jeonju, South Korea
| | - Junghyun Lim
- School of Pharmacy, Jeonbuk National University, Jeonju, South Korea
| | - Soonok Sa
- Food Biotech R&D Center, Samyang Corp., Seongnam-si, South Korea
| | - Go Eun Kim
- Food Biotech R&D Center, Samyang Corp., Seongnam-si, South Korea
| | - Jung Sook Han
- Food Biotech R&D Center, Samyang Corp., Seongnam-si, South Korea
| | - Junghyun Kim
- Department of Oral Pathology, School of Dentistry, Jeonbuk National University, Jeonju, South Korea
| | - Han-Jung Chae
- Non-Clinical Evaluation Center Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, South Korea.,School of Pharmacy, Jeonbuk National University, Jeonju, South Korea
| |
Collapse
|
4
|
Norris D, Yang P, Shin SY, Kearney AL, Kim HJ, Geddes T, Senior AM, Fazakerley DJ, Nguyen LK, James DE, Burchfield JG. Signaling Heterogeneity is Defined by Pathway Architecture and Intercellular Variability in Protein Expression. iScience 2021; 24:102118. [PMID: 33659881 PMCID: PMC7892930 DOI: 10.1016/j.isci.2021.102118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/07/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Insulin's activation of PI3K/Akt signaling, stimulates glucose uptake by enhancing delivery of GLUT4 to the cell surface. Here we examined the origins of intercellular heterogeneity in insulin signaling. Akt activation alone accounted for ~25% of the variance in GLUT4, indicating that additional sources of variance exist. The Akt and GLUT4 responses were highly reproducible within the same cell, suggesting the variance is between cells (extrinsic) and not within cells (intrinsic). Generalized mechanistic models (supported by experimental observations) demonstrated that the correlation between the steady-state levels of two measured signaling processes decreases with increasing distance from each other and that intercellular variation in protein expression (as an example of extrinsic variance) is sufficient to account for the variance in and between Akt and GLUT4. Thus, the response of a population to insulin signaling is underpinned by considerable single-cell heterogeneity that is largely driven by variance in gene/protein expression between cells. Insulin signaling is heterogeneous between cells in the same population The temporal response of signaling components within a cell is highly reproducible Upstream responses (Akt) can only partially predict downstream response (GLUT4) Protein expression variance is a driver of intercellular signaling heterogeneity
Collapse
Affiliation(s)
- Dougall Norris
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Pengyi Yang
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Mathematics and Statistics, The University of Sydney, Sydney, NSW 2006, Australia.,Computational Systems Biology Group, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Sung-Young Shin
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia.,Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Alison L Kearney
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Hani Jieun Kim
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Mathematics and Statistics, The University of Sydney, Sydney, NSW 2006, Australia.,Computational Systems Biology Group, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Thomas Geddes
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Mathematics and Statistics, The University of Sydney, Sydney, NSW 2006, Australia.,Computational Systems Biology Group, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Alistair M Senior
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Daniel J Fazakerley
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Lan K Nguyen
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia.,Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - David E James
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - James G Burchfield
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
5
|
Brumfield A, Chaudhary N, Molle D, Wen J, Graumann J, McGraw TE. Insulin-promoted mobilization of GLUT4 from a perinuclear storage site requires RAB10. Mol Biol Cell 2021; 32:57-73. [PMID: 33175605 PMCID: PMC8098823 DOI: 10.1091/mbc.e20-06-0356] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/30/2020] [Accepted: 11/04/2020] [Indexed: 12/05/2022] Open
Abstract
Insulin controls glucose uptake into muscle and fat cells by inducing a net redistribution of glucose transporter 4 (GLUT4) from intracellular storage to the plasma membrane (PM). The TBC1D4-RAB10 signaling module is required for insulin-stimulated GLUT4 translocation to the PM, although where it intersects GLUT4 traffic was unknown. Here we demonstrate that TBC1D4-RAB10 functions to control GLUT4 mobilization from a trans-Golgi network (TGN) storage compartment, establishing that insulin, in addition to regulating the PM proximal effects of GLUT4-containing vesicles docking to and fusion with the PM, also directly regulates the behavior of GLUT4 deeper within the cell. We also show that GLUT4 is retained in an element/domain of the TGN from which newly synthesized lysosomal proteins are targeted to the late endosomes and the ATP7A copper transporter is translocated to the PM by elevated copper. Insulin does not mobilize ATP7A nor does copper mobilize GLUT4, and RAB10 is not required for copper-elicited ATP7A mobilization. Consequently, GLUT4 intracellular sequestration and mobilization by insulin is achieved, in part, through utilizing a region of the TGN devoted to specialized cargo transport in general rather than being specific for GLUT4. Our results define the GLUT4-containing region of the TGN as a sorting and storage site from which different cargo are mobilized by distinct signals through unique molecular machinery.
Collapse
Affiliation(s)
| | - Natasha Chaudhary
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - Dorothee Molle
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - Jennifer Wen
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - Johannes Graumann
- Weill Cornell Medical College in Qatar, Education City, 24144 Doha, State of Qatar
| | - Timothy E. McGraw
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
- Department of Cardiothoracic Surgery, Weill Cornell Medical College, New York, NY 10065
| |
Collapse
|
6
|
Mu RF, Niu YF, Wang Q, Zhou HM, Hu J, Qin WY, Xiong WY. Eriocalyxin B Inhibits Adipogenesis in 3T3-L1 Adipocytes by Cell Cycle Arrest. NATURAL PRODUCTS AND BIOPROSPECTING 2020; 10:131-140. [PMID: 32314168 PMCID: PMC7253553 DOI: 10.1007/s13659-020-00240-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 04/04/2020] [Indexed: 05/10/2023]
Abstract
Eriocalyxin B, an ent-Kaurene diterpenoid extracted from a traditional Chinese herb Isodon eriocalyx, has been shown to possess multifunctional activities such as anti-cancer and anti-inflammatory. However, the function and mechanism of the compound in adipocyte differentiation is still unknown. Here we reported that eriocalyxin B blunted adipogenesis remarkably by inhibiting the accumulation of lipid droplets, triglycerides and the expressions of adipogenesis-related factors, including C/EBPβ, C/EBPα, PPARγ, and FABP4. Moreover, we showed that the inhibition might be the consequence of cell cycle being arrested at the G2/M phase during the mitotic clonal expansion of adipocyte differentiation, most likely by suppressing mRNAs and proteins of CDK1, CDK2, Cyclin A and Cyclin B1. Overall, we conclude that eriocalyxin B is capable of inhibiting adipocyte differentiation at the early stage through downregulating the proteins involved in cell cycle progression.
Collapse
Affiliation(s)
- Rong-Fang Mu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 Yunnan People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049 People’s Republic of China
| | - Yan-Fen Niu
- Biomedical Engineering Research Center, Kunming Medical University, Kunming, 650500 Yunnan People’s Republic of China
| | - Qian Wang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 Yunnan People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049 People’s Republic of China
| | - Hui-Min Zhou
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 Yunnan People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049 People’s Republic of China
| | - Jing Hu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 Yunnan People’s Republic of China
| | - Wan-Ying Qin
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 Yunnan People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049 People’s Republic of China
| | - Wen-Yong Xiong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 Yunnan People’s Republic of China
- Key Laboratory of Medicinal Chemistry for Natural Resource, Yunnan University, Kunming, 650500 Yunnan People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049 People’s Republic of China
| |
Collapse
|
7
|
Morris S, Geoghegan ND, Sadler JBA, Koester AM, Black HL, Laub M, Miller L, Heffernan L, Simpson JC, Mastick CC, Cooper J, Gadegaard N, Bryant NJ, Gould GW. Characterisation of GLUT4 trafficking in HeLa cells: comparable kinetics and orthologous trafficking mechanisms to 3T3-L1 adipocytes. PeerJ 2020; 8:e8751. [PMID: 32185116 PMCID: PMC7060922 DOI: 10.7717/peerj.8751] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/14/2020] [Indexed: 12/18/2022] Open
Abstract
Insulin-stimulated glucose transport is a characteristic property of adipocytes and muscle cells and involves the regulated delivery of glucose transporter (GLUT4)-containing vesicles from intracellular stores to the cell surface. Fusion of these vesicles results in increased numbers of GLUT4 molecules at the cell surface. In an attempt to overcome some of the limitations associated with both primary and cultured adipocytes, we expressed an epitope- and GFP-tagged version of GLUT4 (HA–GLUT4–GFP) in HeLa cells. Here we report the characterisation of this system compared to 3T3-L1 adipocytes. We show that insulin promotes translocation of HA–GLUT4–GFP to the surface of both cell types with similar kinetics using orthologous trafficking machinery. While the magnitude of the insulin-stimulated translocation of GLUT4 is smaller than mouse 3T3-L1 adipocytes, HeLa cells offer a useful, experimentally tractable, human model system. Here, we exemplify their utility through a small-scale siRNA screen to identify GOSR1 and YKT6 as potential novel regulators of GLUT4 trafficking in human cells.
Collapse
Affiliation(s)
- Silke Morris
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, UK
| | | | - Jessica B A Sadler
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, UK
| | - Anna M Koester
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, UK
| | | | - Marco Laub
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, UK
| | - Lucy Miller
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, UK
| | - Linda Heffernan
- School of Biology & Environmental Science, University College Dublin, Dublin, Ireland
| | - Jeremy C Simpson
- School of Biology & Environmental Science, University College Dublin, Dublin, Ireland
| | | | - Jon Cooper
- School of Engineering, University of Glasgow, Glasgow, UK
| | | | - Nia J Bryant
- Department of Biology, University of York, York, UK
| | - Gwyn W Gould
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
8
|
Lu Y, Ma X, Kong Q, Xu Y, Hu J, Wang F, Qin W, Wang L, Xiong W. Novel dual-color drug screening model for GLUT4 translocation in adipocytes. Mol Cell Probes 2019; 43:6-12. [PMID: 30639558 DOI: 10.1016/j.mcp.2019.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/09/2019] [Accepted: 01/09/2019] [Indexed: 12/22/2022]
Abstract
Insulin-responsive glucose transporter type 4 (GLUT4) translocation plays a major role in controlling glucose uptake in adipose tissue and muscle, maintaining homeostasis and preventing hyperglycemia. Screening for chemicals enhancing GLUT4 translocation is an approach for identifying hits of drug development for type 2 diabetes. Here we developed a novel functional dual-color probe, pHluorin-GLUT4-mOrange2, and constructed 3T3-L1 adipocytes based screening system to simply and efficiently screen new compounds stimulating GLUT4 translocation. Based on this system, we successfully identified a few hits facilitating GLUT4 translocation. In conclusion, we developed an easy-to-apply dual color GLUT4 probe to monitor GLUT4 translocation in insulin-responsive cells, which could be alternatively employed to high-throughput screen compounds regulating GLUT4 translocation and glucose uptake, even to dissect GLTU4 approaching, docking and fusion with the plasma membrane (PM), and to reveal relevant molecular mechanisms involved in these steps as expected.
Collapse
Affiliation(s)
- Yanting Lu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiuli Ma
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Qinghua Kong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Yuhui Xu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Jing Hu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Fang Wang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Wanying Qin
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Libin Wang
- The General Hospital of Ningxia Medical University, Department of Beijing National Biochip Research Center Sub-Center in Ningxia, Yinchuan, 750004, China.
| | - Wenyong Xiong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; The General Hospital of Ningxia Medical University, Department of Beijing National Biochip Research Center Sub-Center in Ningxia, Yinchuan, 750004, China.
| |
Collapse
|
9
|
Hu J, Li X, Tian W, Lu Y, Xu Y, Wang F, Qin W, Ma X, Puno PT, Xiong W. Adenanthin, a Natural ent-Kaurane Diterpenoid Isolated from the Herb Isodon adenantha Inhibits Adipogenesis and the Development of Obesity by Regulation of ROS. Molecules 2019; 24:molecules24010158. [PMID: 30609810 PMCID: PMC6337096 DOI: 10.3390/molecules24010158] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 12/26/2018] [Accepted: 12/27/2018] [Indexed: 02/07/2023] Open
Abstract
Adenanthin, a natural ent-kaurane diterpenoid extracted from the herb Isodon adenantha, has been reported to increase intracellular reactive oxygen species in leukemic and hepatocellular carcinoma cells. However, the function and mechanism of the compound in adipogenesis and the development of obesity is still unknown. In this study, we demonstrated that adenanthin inhibited adipogenesis of 3T3-L1 and mouse embryonic fibroblasts, and the underlying mechanism included two processes: a delayed mitotic clonal expansion via G0/G1 cell cycle arrest by inhibiting the RB-E2F1 signaling pathway and a reduced C/EBPβ signaling by inhibiting the expression and activity of C/EBPβ during mitotic clonal expansion. Furthermore, adenanthin significantly reduced the growing body weight and adipose tissue mass during high-fat diet-inducing obesity of mice, indicating the beneficial effects of adenanthin as a potential agent for prevention of obesity.
Collapse
Affiliation(s)
- Jing Hu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China.
- University of the Chinese Academy of Sciences, Beijing 100049, China.
- Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, China.
| | - Xingren Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China.
- University of the Chinese Academy of Sciences, Beijing 100049, China.
| | - Weifeng Tian
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China.
- University of the Chinese Academy of Sciences, Beijing 100049, China.
| | - Yanting Lu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China.
- University of the Chinese Academy of Sciences, Beijing 100049, China.
| | - Yuhui Xu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China.
- University of the Chinese Academy of Sciences, Beijing 100049, China.
- Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, China.
| | - Fang Wang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China.
- University of the Chinese Academy of Sciences, Beijing 100049, China.
- Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, China.
| | - Wanying Qin
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China.
- University of the Chinese Academy of Sciences, Beijing 100049, China.
| | - Xiuli Ma
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China.
- University of the Chinese Academy of Sciences, Beijing 100049, China.
| | - Pema-Tenzin Puno
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China.
- Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, China.
| | - Wenyong Xiong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China.
- Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, China.
| |
Collapse
|
10
|
McCormick CD, Waters HN, Bezrukov L, Taginya R, Parikh V, Onyekaba GI, Levine JA, Demidowich AP, Yanovski JA, Blank PS, Zimmerberg J. Subcutaneous adipose tissue imaging of human obesity reveals two types of adipocyte membranes: Insulin-responsive and -nonresponsive. J Biol Chem 2018; 293:14249-14259. [PMID: 30006347 DOI: 10.1074/jbc.ra118.003751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/28/2018] [Indexed: 11/06/2022] Open
Abstract
In adipose tissue, resistance to insulin's ability to increase glucose uptake can be induced by multiple factors, including obesity. Impaired insulin action may take place at different spatial loci at the cellular or subcellular level. To begin to understand the spatial response to insulin in human subcutaneous adipose tissue (hSAT), we developed a quantitative imaging method for activation of a major signaling node in the glucoregulatory insulin signaling pathway. After treatment with insulin or control media, biopsied tissues were immunostained for Akt phosphorylation at Thr-308/9 (pAkt) and then imaged by confocal fluorescence microscopy automated to collect a large grid of high resolution fields. In hSAT from 40 men and women with obesity, substantial heterogeneity of pAkt densities in adipocyte membranes were quantified in each image mosaic using a spatial unit of at least twice the size of the point spread function. Statistical analysis of the distribution of pAkt spatial units was best fit as the weighted sum of two separate distributions, corresponding to either a low or high pAkt density. A "high pAkt fraction" metric was calculated from the fraction of high pAkt distributed units over the total units. Importantly, upon insulin stimulation, tissues from the same biopsy showed either a minimal or a substantial change in the high pAkt fraction. Further supporting a two-state response to insulin stimulation, subjects with similar insulin sensitivity indices are also segregated into either of two clusters identified by the amount of membrane-localized pAkt.
Collapse
Affiliation(s)
| | | | | | | | - Viraj Parikh
- Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Ginikanwa I Onyekaba
- Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Jordan A Levine
- Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Andrew P Demidowich
- Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Jack A Yanovski
- Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | | | | |
Collapse
|
11
|
Bae UJ, Jung ES, Jung SJ, Chae SW, Park BH. Mulberry leaf extract displays antidiabetic activity in db/db mice via Akt and AMP-activated protein kinase phosphorylation. Food Nutr Res 2018; 62:1473. [PMID: 30150922 PMCID: PMC6109265 DOI: 10.29219/fnr.v62.1473] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/14/2018] [Accepted: 07/14/2018] [Indexed: 01/05/2023] Open
Abstract
Background Augmenting glucose utilization in skeletal muscle via the phosphatidylinositol-3 kinase (PI3 kinase)/protein kinase B (Akt) pathway or the adenosine monophosphate (AMP)-activated protein kinase (AMPK) pathway is necessary to regulate hyperglycemia in patients with type 2 diabetes mellitus. Objective We investigated the effect of mulberry leaf extract (MLE) on glucose uptake in skeletal muscle cells and explored its in vivo antidiabetic potential. Design Male db/db mice were treated with either MLE (50 mg/kg, 100 mg/kg, and 250 mg/kg) or metformin (100 mg/kg) for 8 weeks. Results MLE treatment stimulated glucose uptake, driven by enhanced translocation of glucose transporter 4 to cell membranes in L6 myotubes. These effects of MLE were synergistic with those of insulin and were abolished in the presence of PI3K inhibitor or AMPK inhibitor. In db/db mice, supplementation with MLE decreased fasting blood glucose and insulin levels and enhanced insulin sensitivity, with increases of p-Akt and p-AMPK in skeletal muscle. Moreover, MLE improved blood lipid parameters and attenuated hepatic steatosis in diabetic db/db mice. Discussion These findings suggest that MLE exerts antidiabetic activity through stimulating glucose disposal in skeletal muscle cells via the PI3K/Akt and AMPK pathways. Conclusions MLE can potentially improve hyperglycemia and hepatic steatosis in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Ui-Jin Bae
- Department of Biochemistry, Chonbuk National University Medical School, Jeonju, Jeonbuk, Republic of Korea.,Clinical Trial Center for Functional Foods, Chonbuk National University Hospital, Jeonju, Jeonbuk, Republic of Korea
| | - Eun-Soo Jung
- Clinical Trial Center for Functional Foods, Chonbuk National University Hospital, Jeonju, Jeonbuk, Republic of Korea
| | - Su-Jin Jung
- Clinical Trial Center for Functional Foods, Chonbuk National University Hospital, Jeonju, Jeonbuk, Republic of Korea
| | - Soo-Wan Chae
- Clinical Trial Center for Functional Foods, Chonbuk National University Hospital, Jeonju, Jeonbuk, Republic of Korea.,Department of Pharmacology, Chonbuk National University Medical School, Jeonju, Jeonbuk, Republic of Korea
| | - Byung-Hyun Park
- Department of Biochemistry, Chonbuk National University Medical School, Jeonju, Jeonbuk, Republic of Korea
| |
Collapse
|
12
|
Tokarz VL, MacDonald PE, Klip A. The cell biology of systemic insulin function. J Cell Biol 2018; 217:2273-2289. [PMID: 29622564 PMCID: PMC6028526 DOI: 10.1083/jcb.201802095] [Citation(s) in RCA: 274] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/21/2018] [Accepted: 03/23/2018] [Indexed: 12/12/2022] Open
Abstract
Insulin is the paramount anabolic hormone, promoting carbon energy deposition in the body. Its synthesis, quality control, delivery, and action are exquisitely regulated by highly orchestrated intracellular mechanisms in different organs or "stations" of its bodily journey. In this Beyond the Cell review, we focus on these five stages of the journey of insulin through the body and the captivating cell biology that underlies the interaction of insulin with each organ. We first analyze insulin's biosynthesis in and export from the β-cells of the pancreas. Next, we focus on its first pass and partial clearance in the liver with its temporality and periodicity linked to secretion. Continuing the journey, we briefly describe insulin's action on the blood vasculature and its still-debated mechanisms of exit from the capillary beds. Once in the parenchymal interstitium of muscle and adipose tissue, insulin promotes glucose uptake into myofibers and adipocytes, and we elaborate on the intricate signaling and vesicle traffic mechanisms that underlie this fundamental function. Finally, we touch upon the renal degradation of insulin to end its action. Cellular discernment of insulin's availability and action should prove critical to understanding its pivotal physiological functions and how their failure leads to diabetes.
Collapse
Affiliation(s)
- Victoria L Tokarz
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Patrick E MacDonald
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
13
|
Chaudhary N, Gonzalez E, Chang SH, Geng F, Rafii S, Altorki NK, McGraw TE. Adenovirus Protein E4-ORF1 Activation of PI3 Kinase Reveals Differential Regulation of Downstream Effector Pathways in Adipocytes. Cell Rep 2017; 17:3305-3318. [PMID: 28009298 DOI: 10.1016/j.celrep.2016.11.082] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 10/05/2016] [Accepted: 11/28/2016] [Indexed: 12/13/2022] Open
Abstract
Insulin activation of phosphatidylinositol 3-kinase (PI3K) regulates metabolism, including the translocation of the Glut4 glucose transporter to the plasma membrane and inactivation of the FoxO1 transcription factor. Adenoviral protein E4-ORF1 stimulates cellular glucose metabolism by mimicking growth-factor activation of PI3K. We have used E4-ORF1 as a tool to dissect PI3K-mediated signaling in adipocytes. E4-ORF1 activation of PI3K in adipocytes recapitulates insulin regulation of FoxO1 but not regulation of Glut4. This uncoupling of PI3K effects occurs despite E4-ORF1 activating PI3K and downstream signaling to levels achieved by insulin. Although E4-ORF1 does not fully recapitulate insulin's effects on Glut4, it enhances insulin-stimulated insertion of Glut4-containing vesicles to the plasma membrane independent of Rab10, a key regulator of Glut4 trafficking. E4-ORF1 also stimulates plasma membrane translocation of ubiquitously expressed Glut1 glucose transporter, an effect that is likely essential for E4-ORF1 to promote an anabolic metabolism in a broad range of cell types.
Collapse
Affiliation(s)
- Natasha Chaudhary
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Eva Gonzalez
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Sung-Hee Chang
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Fuqiang Geng
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Shahin Rafii
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Nasser K Altorki
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY 10065, USA; Lung Cancer Program, Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Timothy E McGraw
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA; Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY 10065, USA; Lung Cancer Program, Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
14
|
Gao L, Chen J, Gao J, Wang H, Xiong W. Super-resolution microscopy reveals the insulin-resistance-regulated reorganization of GLUT4 on plasma membranes. J Cell Sci 2016; 130:396-405. [PMID: 27888215 DOI: 10.1242/jcs.192450] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 11/11/2016] [Indexed: 02/01/2023] Open
Abstract
GLUT4 (also known as SLC2A4) is essential for glucose uptake in skeletal muscles and adipocytes, which play central roles in whole-body glucose metabolism. Here, using direct stochastic optical reconstruction microscopy (dSTORM) to investigate the characteristics of plasma-membrane-fused GLUT4 at the single-molecule level, we have demonstrated that insulin and insulin resistance regulate the spatial organization of GLUT4 in adipocytes. Stimulation with insulin shifted the balance of GLUT4 on the plasma membrane toward a more dispersed configuration. In contrast, insulin resistance induced a more clustered distribution of GLUT4 and increased the mean number of molecules per cluster. Furthermore, our data demonstrate that the F5QQI motif and lipid rafts mediate the maintenance of GLUT4 clusters on the plasma membrane. Mutation of F5QQI (F5QQA-GLUT4) induced a more clustered distribution of GLUT4; moreover, destruction of lipid rafts in adipocytes expressing F5QQA-GLUT4 dramatically decreased the percentage of large clusters and the mean number of molecules per cluster. In conclusion, our data clarify the effects of insulin stimulation or insulin resistance on GLUT4 reorganization on the plasma membrane and reveal new pathogenic mechanisms of insulin resistance.
Collapse
Affiliation(s)
- Lan Gao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, P.R. China.,Graduate University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Junling Chen
- Graduate University of Chinese Academy of Sciences, Beijing 100049, P.R. China.,State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilil 130022, P.R. China
| | - Jing Gao
- Graduate University of Chinese Academy of Sciences, Beijing 100049, P.R. China.,State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilil 130022, P.R. China
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilil 130022, P.R. China
| | - Wenyong Xiong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, P.R. China
| |
Collapse
|
15
|
Bruno J, Brumfield A, Chaudhary N, Iaea D, McGraw TE. SEC16A is a RAB10 effector required for insulin-stimulated GLUT4 trafficking in adipocytes. J Cell Biol 2016; 214:61-76. [PMID: 27354378 PMCID: PMC4932369 DOI: 10.1083/jcb.201509052] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 06/08/2016] [Indexed: 12/19/2022] Open
Abstract
Sec16A is known to be required for COPII vesicle formation from the ER. Here, Bruno et al. show that, independent of its role at the ER, Sec16A is a RAB10 effector involved in the insulin-stimulated formation of specialized transport vesicles that ferry the GLUT4 glucose transporter to the plasma membrane of adipocytes. RAB10 is a regulator of insulin-stimulated translocation of the GLUT4 glucose transporter to the plasma membrane (PM) of adipocytes, which is essential for whole-body glucose homeostasis. We establish SEC16A as a novel RAB10 effector in this process. Colocalization of SEC16A with RAB10 is augmented by insulin stimulation, and SEC16A knockdown attenuates insulin-induced GLUT4 translocation, phenocopying RAB10 knockdown. We show that SEC16A and RAB10 promote insulin-stimulated mobilization of GLUT4 from a perinuclear recycling endosome/TGN compartment. We propose RAB10–SEC16A functions to accelerate formation of the vesicles that ferry GLUT4 to the PM during insulin stimulation. Because GLUT4 continually cycles between the PM and intracellular compartments, the maintenance of elevated cell-surface GLUT4 in the presence of insulin requires accelerated biogenesis of the specialized GLUT4 transport vesicles. The function of SEC16A in GLUT4 trafficking is independent of its previously characterized activity in ER exit site formation and therefore independent of canonical COPII-coated vesicle function. However, our data support a role for SEC23A, but not the other COPII components SEC13, SEC23B, and SEC31, in the insulin stimulation of GLUT4 trafficking, suggesting that vesicles derived from subcomplexes of COPII coat proteins have a role in the specialized trafficking of GLUT4.
Collapse
Affiliation(s)
- Joanne Bruno
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065 Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10065
| | | | - Natasha Chaudhary
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - David Iaea
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - Timothy E McGraw
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065 Department of Cardiothoracic Surgery, Weill Cornell Medical College, New York, NY 10065
| |
Collapse
|
16
|
Fan R, Cheng RR, Zhu HT, Wang D, Yang CR, Xu M, Zhang YJ. Two New Oleanane-type Triterpenoids from Methanolyzed Saponins of Momordica cochinchinensis. Nat Prod Commun 2016. [DOI: 10.1177/1934578x1601100607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Two new oleanane-type triterpenoid saponins (1 and 2) were isolated from the methanolyzed total saponins of the seeds of Momordica cochinchinensis (Lour.) Spreng, together with 16 known compounds (3—18). Their structures were elucidated on the basis of detailed spectroscopic, including ID and 2D NMR, mass spectrometric, methanolysis and LC-MS analysis. All the isolates were tested for their cytotoxic activities against five human cancer cell lines (HL-60, SMMC-7721, PANC-1, A-549, and SW-480) and the glucose uptake activity. The known compound 6 exhibited toxic effects against HL-60 with an IC50 value of 18.1 μM, while 10 showed cytotoxicity against SMMC-7721 and A-549 cell lines, with IC50 values of 34.4 and 32.8 μM, respectively. In addition, the new compound 2 showed glucose uptake activity with a glucose consumption value of 0.29 μM at 10 μM concentration.
Collapse
Affiliation(s)
- Rong Fan
- State Key Laboratory of Phytochemistry and Plant Resources of West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, P. R. China
- Yunnan University of Traditional Chinese Medicine, Kunming, 650500, P. R. China
| | - Rong-Rong Cheng
- State Key Laboratory of Phytochemistry and Plant Resources of West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, P. R. China
| | - Hong-Tao Zhu
- State Key Laboratory of Phytochemistry and Plant Resources of West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, P. R. China
| | - Dong Wang
- State Key Laboratory of Phytochemistry and Plant Resources of West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, P. R. China
| | - Chong-Ren Yang
- State Key Laboratory of Phytochemistry and Plant Resources of West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, P. R. China
| | - Min Xu
- State Key Laboratory of Phytochemistry and Plant Resources of West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, P. R. China
| | - Ying-Jun Zhang
- State Key Laboratory of Phytochemistry and Plant Resources of West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, P. R. China
| |
Collapse
|
17
|
Gongpan P, Lu Y, Wang F, Xu Y, Xiong W. AS160 controls eukaryotic cell cycle and proliferation by regulating the CDK inhibitor p21. Cell Cycle 2016; 15:1733-41. [PMID: 27152871 DOI: 10.1080/15384101.2016.1183853] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
AS160 (TBC1D4) has been implicated in multiple biological processes. However, the role and the mechanism of action of AS160 in the regulation of cell proliferation remain unclear. In this study, we demonstrated that AS160 knockdown led to blunted cell proliferation in multiple cell types, including fibroblasts and cancer cells. The results of cell cycle analysis showed that these cells were arrested in the G1 phase. Intriguingly, this inhibition of cell proliferation and the cell cycle arrest caused by AS160 depletion were glucose independent. Moreover, AS160 silencing led to a marked upregulation of the expression of the cyclin-dependent kinase inhibitor p21. Furthermore, whereas AS160 overexpression resulted in p21 downregulation and rescued the arrested cell cycle in AS160-depeleted cells, p21 silencing rescued the inhibited cell cycle and proliferation in the cells. Thus, our results demonstrated that AS160 regulates glucose-independent eukaryotic cell proliferation through p21-dependent control of the cell cycle, and thereby revealed a molecular mechanism of AS160 modulation of cell cycle and proliferation that is of general physiological significance.
Collapse
Affiliation(s)
- Pianchou Gongpan
- a State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany , Chinese Academy of Sciences , Kunming , Yunnan , P.R. China.,b Graduate University of Chinese Academy of Sciences , Beijing , P.R. China
| | - Yanting Lu
- a State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany , Chinese Academy of Sciences , Kunming , Yunnan , P.R. China.,b Graduate University of Chinese Academy of Sciences , Beijing , P.R. China
| | - Fang Wang
- a State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany , Chinese Academy of Sciences , Kunming , Yunnan , P.R. China.,b Graduate University of Chinese Academy of Sciences , Beijing , P.R. China
| | - Yuhui Xu
- a State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany , Chinese Academy of Sciences , Kunming , Yunnan , P.R. China.,b Graduate University of Chinese Academy of Sciences , Beijing , P.R. China
| | - Wenyong Xiong
- a State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany , Chinese Academy of Sciences , Kunming , Yunnan , P.R. China
| |
Collapse
|
18
|
Talantikite M, Berenguer M, Gonzalez T, Alessi MC, Poggi M, Peiretti F, Govers R. The first intracellular loop of GLUT4 contains a retention motif. J Cell Sci 2016; 129:2273-84. [DOI: 10.1242/jcs.183525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 04/20/2016] [Indexed: 01/02/2023] Open
Abstract
Glucose transporter GLUT4 plays a major role in glucose homeostasis and is efficiently retained intracellularly in adipocytes and myocytes. To simplify the analysis of its retention, various intracellular GLUT4 domains were fused individually to reporter molecules. Of the four short cytoplasmic loops of GLUT4, only the first nine-residue-long loop conferred intracellular retention of truncated forms of the transferrin receptor and CD4 in adipocytes. In contrast, the same loop of GLUT1 was without effect. The reporter molecules to which the first loop of GLUT4 was fused localized, unlike GLUT4, to the TGN, possibly explaining why these molecules did not respond to insulin. The retention induced by the GLUT4 loop was specific to adipocytes as it did not induce retention in preadipocytes. Of the SQWLGRKRA sequence that constitutes this loop, mutation of either the tryptophan or lysine residue abrogated reporter retention. Mutation of these residues individually into alanines in the full-length GLUT4 molecule resulted in a decreased retention for GLUT4-W105A. We conclude that the first intracellular loop of GLUT4 contains retention motif WLGRK, in which Trp105 plays a prominent role.
Collapse
Affiliation(s)
- Maya Talantikite
- Inserm U1062, INRA1260, Aix Marseille University, Faculty of Medicine, Marseille F-13385, France
| | - Marion Berenguer
- Inserm U895, Mediterranean Research Center for Molecular Medicine (C3M), Nice, F-06204, France
| | - Teresa Gonzalez
- Inserm U1062, INRA1260, Aix Marseille University, Faculty of Medicine, Marseille F-13385, France
| | - Marie Christine Alessi
- Inserm U1062, INRA1260, Aix Marseille University, Faculty of Medicine, Marseille F-13385, France
| | - Marjorie Poggi
- Inserm U1062, INRA1260, Aix Marseille University, Faculty of Medicine, Marseille F-13385, France
| | - Franck Peiretti
- Inserm U1062, INRA1260, Aix Marseille University, Faculty of Medicine, Marseille F-13385, France
| | - Roland Govers
- Inserm U1062, INRA1260, Aix Marseille University, Faculty of Medicine, Marseille F-13385, France
- Inserm U895, Mediterranean Research Center for Molecular Medicine (C3M), Nice, F-06204, France
| |
Collapse
|
19
|
Tolvanen TA, Dash SN, Polianskyte-Prause Z, Dumont V, Lehtonen S. Lack of CD2AP disrupts Glut4 trafficking and attenuates glucose uptake in podocytes. J Cell Sci 2015; 128:4588-600. [PMID: 26546360 DOI: 10.1242/jcs.175075] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 11/02/2015] [Indexed: 12/13/2022] Open
Abstract
The adapter protein CD2-associated protein (CD2AP) functions in various signaling and vesicle trafficking pathways, including endosomal sorting and/or trafficking and degradation pathways. Here, we investigated the role of CD2AP in insulin-dependent glucose transporter 4 (Glut4, also known as SLC2A4) trafficking and glucose uptake. Glucose uptake was attenuated in CD2AP(-/-) podocytes compared with wild-type podocytes in the basal state, and CD2AP(-/-) podocytes failed to increase glucose uptake in response to insulin. Live-cell imaging revealed dynamic trafficking of HA-Glut4-GFP in wild-type podocytes, whereas in CD2AP(-/-) podocytes, HA-Glut4-GFP clustered perinuclearly. In subcellular membrane fractionations, CD2AP co-fractionated with Glut4, IRAP (also known as LNPEP) and sortilin, constituents of Glut4 storage vesicles (GSVs). We further found that CD2AP forms a complex with GGA2, a clathrin adaptor, which sorts Glut4 to GSVs, suggesting a role for CD2AP in this process. We also found that CD2AP forms a complex with clathrin and connects clathrin to actin in the perinuclear region. Furthermore, clathrin recycling back to trans-Golgi membranes from the vesicular fraction containing GSVs was defective in the absence of CD2AP. This leads to reduced insulin-stimulated trafficking of GSVs and attenuated glucose uptake into CD2AP(-/-) podocytes.
Collapse
Affiliation(s)
- Tuomas A Tolvanen
- Department of Pathology, University of Helsinki, 00290 Helsinki, Finland
| | | | | | - Vincent Dumont
- Department of Pathology, University of Helsinki, 00290 Helsinki, Finland
| | - Sanna Lehtonen
- Department of Pathology, University of Helsinki, 00290 Helsinki, Finland
| |
Collapse
|
20
|
Lanzerstorfer P, Stadlbauer V, Chtcheglova LA, Haselgrübler R, Borgmann D, Wruss J, Hinterdorfer P, Schröder K, Winkler SM, Höglinger O, Weghuber J. Identification of novel insulin mimetic drugs by quantitative total internal reflection fluorescence (TIRF) microscopy. Br J Pharmacol 2015; 171:5237-51. [PMID: 25039620 PMCID: PMC4262000 DOI: 10.1111/bph.12845] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 06/18/2014] [Accepted: 06/27/2014] [Indexed: 12/25/2022] Open
Abstract
Background and Purpose Insulin stimulates the transport of glucose in target tissues by triggering the translocation of glucose transporter 4 (GLUT4) to the plasma membrane. Resistance to insulin, the major abnormality in type 2 diabetes, results in a decreased GLUT4 translocation efficiency. Thus, special attention is being paid to search for compounds that are able to enhance this translocation process in the absence of insulin. Experimental Approach Total internal reflection fluorescence (TIRF) microscopy was applied to quantify GLUT4 translocation in highly insulin-sensitive CHO-K1 cells expressing a GLUT4-myc-GFP fusion protein. Key Results Using our approach, we demonstrated GLUT4 translocation modulatory properties of selected substances and identified novel potential insulin mimetics. An increase in the TIRF signal was found to correlate with an elevated glucose uptake. Variations in the expression level of the human insulin receptor (hInsR) showed that the insulin mimetics identified stimulate GLUT4 translocation by a mechanism that is independent of the presence of the hInsR. Conclusions and Implications Taken together, the results indicate that TIRF microscopy is an excellent tool for the quantification of GLUT4 translocation and for identifying insulin mimetic drugs.
Collapse
Affiliation(s)
- Peter Lanzerstorfer
- School of Engineering and Environmental Sciences, University of Applied Sciences Upper Austria, Wels, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Zhou M, Zhou K, Gao XM, Jiang ZY, Lv JJ, Liu ZH, Yang GY, Miao MM, Che CT, Hu QF. Fistulains A and B, New Bischromones from the Bark of Cassia fistula, and Their Activities. Org Lett 2015; 17:2638-41. [PMID: 25965347 DOI: 10.1021/acs.orglett.5b01007] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Fistulains A and B (1 and 2), two novel bischromones with unique coupling patterns, alone with their biosynthetic related compound 3, were isolated from the bark of Cassia fistula. Fistulain A represents a new type of dimeric chromone alkaloid biogenetically derived from a chromone and a tricyclic alkaloid through an unusual C-14-N linkage. Fistulain B has a new carbon skeleton with a C-14-C-5' linkage formed between two different chromone units. Fistulain A displayed anti-TMV activity, and both 1 and 2 showed weak cytotoxicities.
Collapse
Affiliation(s)
- Min Zhou
- †Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission and Ministry of Education, Yunnan Minzu University, Kunming 650031, Yunnan, People's Republic of China.,‡Key Laboratory of Tobacco Chemistry of Yunnan Province, China Tobacco Yunnan Industrial Co., Ltd, Kunming 650231, Yunnan, People's Republic of China
| | - Kun Zhou
- †Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission and Ministry of Education, Yunnan Minzu University, Kunming 650031, Yunnan, People's Republic of China
| | - Xue-Mei Gao
- †Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission and Ministry of Education, Yunnan Minzu University, Kunming 650031, Yunnan, People's Republic of China
| | - Zhi-Yong Jiang
- †Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission and Ministry of Education, Yunnan Minzu University, Kunming 650031, Yunnan, People's Republic of China
| | - Jun-Jiang Lv
- ∥Chemistry and Chemical Engineering College, Chongqing University, Chongqing 400030, People's Republic of China
| | - Zhi-Hua Liu
- ‡Key Laboratory of Tobacco Chemistry of Yunnan Province, China Tobacco Yunnan Industrial Co., Ltd, Kunming 650231, Yunnan, People's Republic of China
| | - Guang-Yu Yang
- ‡Key Laboratory of Tobacco Chemistry of Yunnan Province, China Tobacco Yunnan Industrial Co., Ltd, Kunming 650231, Yunnan, People's Republic of China
| | - Ming-Ming Miao
- ‡Key Laboratory of Tobacco Chemistry of Yunnan Province, China Tobacco Yunnan Industrial Co., Ltd, Kunming 650231, Yunnan, People's Republic of China
| | - Chun-Tao Che
- §Department of Medicinal Chemistry and Pharmacognosy and WHO Collaborating Center for Traditional Medicine, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Qiu-Fen Hu
- †Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission and Ministry of Education, Yunnan Minzu University, Kunming 650031, Yunnan, People's Republic of China.,§Department of Medicinal Chemistry and Pharmacognosy and WHO Collaborating Center for Traditional Medicine, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| |
Collapse
|
22
|
Tian LW, Tao MK, Xu M, Hu J, Zhu HT, Xiong WY, Wang D, Yang CR, Zhang YJ. Carboxymethyl- and carboxyl-catechins from ripe Pu-er tea. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:12229-12234. [PMID: 25455197 DOI: 10.1021/jf5036959] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Ripe Pu-er tea, a special microbial postfermented tea originated from Yunnan Province, China, since ancient times, is made from green Pu-er tea prepared from the leaves of Camellia sinensis var. assamica (Theaceae). Chemical investigation on thearubigin (n-BuOH-soluble) fraction of the commercial ripe Pu-er tea, led to the identification of four new flavan-3-ol derivatives, 8-carboxymethyl-(+)-catechin (1), 8-carboxymethyl-(+)-catechin methyl ester (2), 6-carboxymethyl-(+)-catechin (3), and 6-carboxyl-(-)-gallocatechin (4), together with 18 known compounds, including other three flavan-3-ol derivatives (5-7), 10 flavonoid glycosides (8-17), two hydrolyzable tannins (18 and 19), two quinic acid derivatives (20-21), and a purine alkaloid (22). Flavonoid glycosides 8-11 are reported from tea plants for the first time. The thearubigin fraction of ripe Pu-er tea was qualitatively analyzed by HPLC, and gallic acid was found to be the major component. Compounds 4, 6-17, 21 and 22 were tested for their acute activities on insulin sensitivity in differentiated 3T3-L1 adipocytes, but none of them showed significant bioactivity at a concentration of 10 μM.
Collapse
Affiliation(s)
- Li-Wen Tian
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences , Kunming 650201, P. R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Govers R. Molecular mechanisms of GLUT4 regulation in adipocytes. DIABETES & METABOLISM 2014; 40:400-10. [PMID: 24656589 DOI: 10.1016/j.diabet.2014.01.005] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 01/24/2014] [Accepted: 01/26/2014] [Indexed: 01/28/2023]
|
24
|
Kioumourtzoglou D, Sadler JBA, Black HL, Berends R, Wellburn C, Bryant NJ, Gould GW. Studies of the regulated assembly of SNARE complexes in adipocytes. Biochem Soc Trans 2014; 42:1396-400. [PMID: 25233421 DOI: 10.1042/bst20140114] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2024]
Abstract
Insulin plays a fundamental role in whole-body glucose homeostasis. Central to this is the hormone's ability to rapidly stimulate the rate of glucose transport into adipocytes and muscle cells [1]. Upon binding its receptor, insulin stimulates an intracellular signalling cascade that culminates in redistribution of glucose transporter proteins, specifically the GLUT4 isoform, from intracellular stores to the plasma membrane, a process termed 'translocation' [1,2]. This is an example of regulated membrane trafficking [3], a process that also underpins other aspects of physiology in a number of specialized cell types, for example neurotransmission in brain/neurons and release of hormone-containing vesicles from specialized secretory cells such as those found in pancreatic islets. These processes invoke a number of intriguing biological questions as follows. How is the machinery involved in these membrane trafficking events mobilized in response to a stimulus? How do the signalling pathways that detect the external stimulus interface with the trafficking machinery? Recent studies of insulin-stimulated GLUT4 translocation offer insight into such questions. In the present paper, we have reviewed these studies and draw parallels with other regulated trafficking systems.
Collapse
Affiliation(s)
- Dimitrios Kioumourtzoglou
- *Henry Wellcome Laboratory of Cell Biology, Institute for Molecular, Cell and Systems Biology, Davidson Building, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K
| | - Jessica B A Sadler
- *Henry Wellcome Laboratory of Cell Biology, Institute for Molecular, Cell and Systems Biology, Davidson Building, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K
| | - Hannah L Black
- †Department of Biology, University of York, Heslington, York YO10 5DD, U.K
| | - Rebecca Berends
- *Henry Wellcome Laboratory of Cell Biology, Institute for Molecular, Cell and Systems Biology, Davidson Building, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K
| | - Cassie Wellburn
- *Henry Wellcome Laboratory of Cell Biology, Institute for Molecular, Cell and Systems Biology, Davidson Building, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K
| | - Nia J Bryant
- †Department of Biology, University of York, Heslington, York YO10 5DD, U.K
| | - Gwyn W Gould
- *Henry Wellcome Laboratory of Cell Biology, Institute for Molecular, Cell and Systems Biology, Davidson Building, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K
| |
Collapse
|
25
|
Tessneer KL, Jackson RM, Griesel BA, Olson AL. Rab5 activity regulates GLUT4 sorting into insulin-responsive and non-insulin-responsive endosomal compartments: a potential mechanism for development of insulin resistance. Endocrinology 2014; 155:3315-28. [PMID: 24932807 PMCID: PMC4138579 DOI: 10.1210/en.2013-2148] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Glucose transporter isoform 4 (GLUT4) is the insulin-responsive glucose transporter mediating glucose uptake in adipose and skeletal muscle. Reduced GLUT4 translocation from intracellular storage compartments to the plasma membrane is a cause of peripheral insulin resistance. Using a chronic hyperinsulinemia (CHI)-induced cell model of insulin resistance and Rab5 mutant overexpression, we determined these manipulations altered endosomal sorting of GLUT4, thus contributing to the development of insulin resistance. We found that CHI induced insulin resistance in 3T3-L1 adipocytes by retaining GLUT4 in a Rab5-activity-dependent compartment that is unable to equilibrate with the cell surface in response to insulin. Furthermore, CHI-mediated retention of GLUT4 in this non-insulin-responsive compartment impaired filling of the transferrin receptor (TfR)-positive and TfR-negative insulin-responsive storage compartments. Our data suggest that hyperinsulinemia may inhibit GLUT4 by chronically maintaining GLUT4 in the Rab5 activity-dependent endosomal pathway and impairing formation of the TfR-negative and TfR-positive insulin-responsive GLUT4 pools. This model suggests that an early event in the development of insulin-resistant glucose transport in adipose tissue is to alter the intracellular localization of GLUT4 to a compartment that does not efficiently equilibrate with the cell surface when insulin levels are elevated for prolonged periods of time.
Collapse
Affiliation(s)
- Kandice L Tessneer
- Department of Biochemistry and Molecular Biology (K.L.T., R.M.J., B.A.G., A.L.O.), University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73126; and Cardiovascular Biology Program (K.L.T.), Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| | | | | | | |
Collapse
|
26
|
Foley KP, Klip A. Dynamic GLUT4 sorting through a syntaxin-6 compartment in muscle cells is derailed by insulin resistance-causing ceramide. Biol Open 2014; 3:314-25. [PMID: 24705014 PMCID: PMC4021353 DOI: 10.1242/bio.20147898] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
GLUT4 constitutively recycles between the plasma membrane and intracellular depots. Insulin shifts this dynamic equilibrium towards the plasma membrane by recruiting GLUT4 to the plasma membrane from insulin-responsive vesicles. Muscle is the primary site for dietary glucose deposition; however, how GLUT4 sorts into insulin-responsive vesicles, and if and how insulin resistance affects this process, is unknown. In L6 myoblasts stably expressing myc-tagged GLUT4, we analyzed the intracellular itinerary of GLUT4 as it internalizes from the cell surface and examined if such sorting is perturbed by C2-ceramide, a lipid metabolite causing insulin resistance. Surface-labeled GLUT4myc that internalized for 30 min accumulated in a Syntaxin-6 (Stx6)- and Stx16-positive perinuclear sub-compartment devoid of furin or internalized transferrin, and displayed insulin-responsive re-exocytosis. C2-ceramide dispersed the Stx6-positive sub-compartment and prevented insulin-responsive re-exocytosis of internalized GLUT4myc, even under conditions not affecting insulin-stimulated signaling towards Akt. Microtubule disruption with nocodazole prevented pre-internalized GLUT4myc from reaching the Stx6-positive perinuclear sub-compartment and from undergoing insulin-responsive exocytosis. Removing nocodazole allowed both parameters to recover, suggesting that the Stx6-positive perinuclear sub-compartment was required for GLUT4 insulin-responsiveness. Accordingly, Stx6 knockdown inhibited by ∼50% the ability of internalized GLUT4myc to undergo insulin-responsive re-exocytosis without altering its overall perinuclear accumulation. We propose that Stx6 defines the insulin-responsive compartment in muscle cells. Our data are consistent with a model where ceramide could cause insulin resistance by altering intracellular GLUT4 sorting.
Collapse
Affiliation(s)
- Kevin P Foley
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Amira Klip
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
27
|
Abstract
GLUT4 is regulated by its intracellular localization. In the absence of insulin, GLUT4 is efficiently retained intracellularly within storage compartments in muscle and fat cells. Upon insulin stimulation (and contraction in muscle), GLUT4 translocates from these compartments to the cell surface where it transports glucose from the extracellular milieu into the cell. Its implication in insulin-regulated glucose uptake makes GLUT4 not only a key player in normal glucose homeostasis but also an important element in insulin resistance and type 2 diabetes. Nevertheless, how GLUT4 is retained intracellularly and how insulin acts on this retention mechanism is largely unclear. In this review, the current knowledge regarding the various molecular processes that govern GLUT4 physiology is discussed as well as the questions that remain.
Collapse
|
28
|
Liang CQ, Hu J, Shi YM, Shang SZ, Du X, Zhan R, Wang WG, Xiong WY, Xiao WL, Zhang HB, Sun HD. Five New Nortriterpenoids from the Stems ofSchisandra neglecta. Helv Chim Acta 2013. [DOI: 10.1002/hlca.201200392] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
29
|
Sadacca LA, Bruno J, Wen J, Xiong W, McGraw TE. Specialized sorting of GLUT4 and its recruitment to the cell surface are independently regulated by distinct Rabs. Mol Biol Cell 2013; 24:2544-57. [PMID: 23804653 PMCID: PMC3744946 DOI: 10.1091/mbc.e13-02-0103] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
RAB10 and RAB14 function at sequential steps of insulin-stimulated GLUT4 translocation to the plasma membrane. RAB14 functions upstream of RAB10 in GLUT4 sorting to the specialized transport vesicles, and RAB10 and its GAP protein comprise the main signaling module that regulates the accumulation of GLUT4 transport vesicles at the plasma membrane. Adipocyte glucose uptake in response to insulin is essential for physiological glucose homeostasis: stimulation of adipocytes with insulin results in insertion of the glucose transporter GLUT4 into the plasma membrane and subsequent glucose uptake. Here we establish that RAB10 and RAB14 are key regulators of GLUT4 trafficking that function at independent, sequential steps of GLUT4 translocation. RAB14 functions upstream of RAB10 in the sorting of GLUT4 to the specialized transport vesicles that ferry GLUT4 to the plasma membrane. RAB10 and its GTPase-activating protein (GAP) AS160 comprise the principal signaling module downstream of insulin receptor activation that regulates the accumulation of GLUT4 transport vesicles at the plasma membrane. Although both RAB10 and RAB14 are regulated by the GAP activity of AS160 in vitro, only RAB10 is under the control of AS160 in vivo. Insulin regulation of the pool of RAB10 required for GLUT4 translocation occurs through regulation of AS160, since activation of RAB10 by DENND4C, its GTP exchange factor, does not require insulin stimulation.
Collapse
Affiliation(s)
- L Amanda Sadacca
- Department of Biochemistry, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | | | | | | | | |
Collapse
|
30
|
Liang CQ, Hu J, Shi YM, Shang SZ, Du X, Zhan R, Xiong WY, Zhang HB, Xiao WL, Sun HD. Schisphenlignans A-E: five new dibenzocyclooctadiene lignans from Schisandra sphenanthera. Chem Pharm Bull (Tokyo) 2013; 61:96-100. [PMID: 23302592 DOI: 10.1248/cpb.c12-00700] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Five new dibenzocyclooctadiene lignans, schisphenlignans A-E (1-5), together with eight known ones, were isolated from the stems of Schisandra sphenanthera. The structures of 1-5 were elucidated based on the analysis of their NMR, MS and circular dichroism (CD) spectra. Some isolates were tested for their acute activities on insulin sensitivity in 3T3-L1 differentiated adipocytes, but none of them showed significant bioactivity with 10 µM administration of the tested compounds.
Collapse
Affiliation(s)
- Cheng-Qin Liang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Six new lignans from the leaves and stems of Schisandra sphenanthera. Fitoterapia 2013; 86:171-7. [PMID: 23500381 DOI: 10.1016/j.fitote.2013.03.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 02/26/2013] [Accepted: 03/03/2013] [Indexed: 11/23/2022]
Abstract
Six new lignans, schisphenlignans F-K (1-6), together with ten known ones, were isolated from the leaves and stems of Schisandra sphenanthera. Their structures were elucidated on the basis of spectroscopic methods, including extensive NMR, MS and CD spectra. In addition, some compounds were tested for their acute activity on insulin sensitivity in 3T3-L1 differentiated adipocytes and anti-HIV-1 activity.
Collapse
|
32
|
Penque BA, Tackett L, Elmendorf JS. Trivalent Chromium Modulates Hexosamine Biosynthesis Pathway Transcriptional Activation of Cholesterol Synthesis and Insulin Resistance. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/ojemd.2013.34a1001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
33
|
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
|
34
|
Haga Y, Ishii K, Hibino K, Sako Y, Ito Y, Taniguchi N, Suzuki T. Visualizing specific protein glycoforms by transmembrane fluorescence resonance energy transfer. Nat Commun 2012; 3:907. [DOI: 10.1038/ncomms1906] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 05/14/2012] [Indexed: 11/09/2022] Open
|
35
|
Li J, Malaby AW, Famulok M, Sabe H, Lambright DG, Hsu VW. Grp1 plays a key role in linking insulin signaling to glut4 recycling. Dev Cell 2012; 22:1286-98. [PMID: 22609160 DOI: 10.1016/j.devcel.2012.03.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 01/31/2012] [Accepted: 03/13/2012] [Indexed: 11/29/2022]
Abstract
The glucose transporter type 4 (glut4) is critical for metabolic homeostasis. Insulin regulates glut4 by modulating its expression on the cell surface. This regulation is mainly achieved by targeting the endocytic recycling of glut4. We identify general receptor for 3-phosphoinositides 1 (Grp1) as a guanine nucleotide exchange factor for ADP-ribosylation factor 6 (ARF6) that promotes glut4 vesicle formation. Grp1 also promotes the later steps of glut4 recycling through ARF6. Insulin signaling regulates Grp1 through phosphorylation by Akt. We also find that mutations that mimic constitutive phosphorylation of Grp1 can bypass upstream insulin signaling to induce glut4 recycling. Thus, we have uncovered a major mechanism by which insulin regulates glut4 recycling. Our findings also reveal the complexity by which a single small GTPase in vesicular transport can coordinate its multiple steps to accomplish a round of transport.
Collapse
Affiliation(s)
- Jian Li
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, and Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
36
|
Habegger KM, Hoffman NJ, Ridenour CM, Brozinick JT, Elmendorf JS. AMPK enhances insulin-stimulated GLUT4 regulation via lowering membrane cholesterol. Endocrinology 2012; 153:2130-41. [PMID: 22434076 PMCID: PMC3339638 DOI: 10.1210/en.2011-2099] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AMP-activated protein kinase (AMPK) enhances glucose transporter GLUT4 regulation. AMPK also suppresses energy-consuming pathways such as cholesterol synthesis. Interestingly, recent in vitro and in vivo data suggest that excess membrane cholesterol impairs GLUT4 regulation. Therefore, this study tested whether a beneficial, GLUT4-regulatory aspect of AMPK stimulation involved cholesterol lowering. Using L6 myotubes stably expressing an exofacial myc-epitope-tagged-GLUT4, AMPK stimulation by 5-aminoimidazole-4-carboxamide-1-β-d-ribonucleoside (AICAR; 45 min, 1 mm) or 2,4-dinitrophenol (DNP; 30 min, 200 μm) increased cell surface GLUT4myc labeling by approximately ≈ 25% (P < 0.05). Insulin (20 min, 100 nm) also increased GLUT4myc labeling by about 50% (P < 0.05), which was further enhanced (≈ 25%, P < 0.05) by AICAR or DNP. Consistent with AMPK-mediated suppression of cholesterol synthesis, AICAR and DNP decreased membrane cholesterol by 20-25% (P < 0.05). Whereas AMPK knockdown prevented the enhanced basal and insulin-stimulated GLUT4myc labeling by AICAR and DNP, cholesterol replenishment only blocked the AMPK-associated enhancement in insulin action. Cells cultured in a hyperinsulinemic milieu, resembling conditions in vivo that promote the progression/worsening of insulin resistance, displayed an increase in membrane cholesterol. This occurred concomitantly with a loss of cortical filamentous actin (F-actin) and defects in GLUT4 regulation by insulin. These derangements were prevented by AMPK stimulation. Examination of skeletal muscle from insulin-resistant Zucker rats revealed a similar elevation in membrane cholesterol and loss of F-actin. Lowering cholesterol to control levels restored F-actin structure and insulin sensitivity. In conclusion, these data suggest a novel aspect of GLUT4 regulation by AMPK involves membrane cholesterol lowering. Moreover, this AMPK-mediated process protected against hyperinsulinemia-induced insulin resistance.
Collapse
Affiliation(s)
- Kirk M Habegger
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | |
Collapse
|
37
|
Habegger KM, Penque BA, Sealls W, Tackett L, Bell LN, Blue EK, Gallagher PJ, Sturek M, Alloosh MA, Steinberg HO, Considine RV, Elmendorf JS. Fat-induced membrane cholesterol accrual provokes cortical filamentous actin destabilisation and glucose transport dysfunction in skeletal muscle. Diabetologia 2012; 55:457-67. [PMID: 22002007 PMCID: PMC3245823 DOI: 10.1007/s00125-011-2334-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Accepted: 09/19/2011] [Indexed: 11/26/2022]
Abstract
AIMS/HYPOTHESIS Diminished cortical filamentous actin (F-actin) has been implicated in skeletal muscle insulin resistance, yet the mechanism(s) is unknown. Here we tested the hypothesis that changes in membrane cholesterol could be a causative factor, as organised F-actin structure emanates from cholesterol-enriched raft microdomains at the plasma membrane. METHODS Skeletal muscle samples from high-fat-fed animals and insulin-sensitive and insulin-resistant human participants were evaluated. The study also used L6 myotubes to directly determine the impact of fatty acids (FAs) on membrane/cytoskeletal variables and insulin action. RESULTS High-fat-fed insulin-resistant animals displayed elevated levels of membrane cholesterol and reduced F-actin structure compared with normal chow-fed animals. Moreover, human muscle biopsies revealed an inverse correlation between membrane cholesterol and whole-body glucose disposal. Palmitate-induced insulin-resistant myotubes displayed membrane cholesterol accrual and F-actin loss. Cholesterol lowering protected against the palmitate-induced defects, whereas characteristically measured defects in insulin signalling were not corrected. Conversely, cholesterol loading of L6 myotube membranes provoked a palmitate-like cytoskeletal/GLUT4 derangement. Mechanistically, we observed a palmitate-induced increase in O-linked glycosylation, an end-product of the hexosamine biosynthesis pathway (HBP). Consistent with HBP activity affecting the transcription of various genes, we observed an increase in Hmgcr, a gene that encodes 3-hydroxy-3-methyl-glutaryl coenzyme A reductase, the rate-limiting enzyme in cholesterol synthesis. In line with increased HBP activity transcriptionally provoking a membrane cholesterol-based insulin-resistant state, HBP inhibition attenuated Hmgcr expression and prevented membrane cholesterol accrual, F-actin loss and GLUT4/glucose transport dysfunction. CONCLUSIONS/INTERPRETATION Our results suggest a novel cholesterolgenic-based mechanism of FA-induced membrane/cytoskeletal disorder and insulin resistance.
Collapse
Affiliation(s)
- K. M. Habegger
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, VanNuys Medical Science Building Rm 308A, 635 Barnhill Drive, Indianapolis, IN 46202-5120 USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN USA
- Indiana University School of Medicine Center for Diabetes Research, Indianapolis, IN USA
- Indiana University School of Medicine Center for Membrane Biosciences, Indianapolis, IN USA
| | - B. A. Penque
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, VanNuys Medical Science Building Rm 308A, 635 Barnhill Drive, Indianapolis, IN 46202-5120 USA
- Indiana University School of Medicine Center for Diabetes Research, Indianapolis, IN USA
- Indiana University School of Medicine Center for Membrane Biosciences, Indianapolis, IN USA
| | - W. Sealls
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, VanNuys Medical Science Building Rm 308A, 635 Barnhill Drive, Indianapolis, IN 46202-5120 USA
- Indiana University School of Medicine Center for Diabetes Research, Indianapolis, IN USA
- Indiana University School of Medicine Center for Membrane Biosciences, Indianapolis, IN USA
- Indiana University School of Medicine Center for Vascular Biology and Medicine, Indianapolis, IN USA
| | - L. Tackett
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, VanNuys Medical Science Building Rm 308A, 635 Barnhill Drive, Indianapolis, IN 46202-5120 USA
| | - L. N. Bell
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, VanNuys Medical Science Building Rm 308A, 635 Barnhill Drive, Indianapolis, IN 46202-5120 USA
- Indiana University School of Medicine Center for Diabetes Research, Indianapolis, IN USA
- Department of Medicine, Division of Endocrinology and Metabolism, Indiana University School of Medicine, Indianapolis, IN USA
| | - E. K. Blue
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, VanNuys Medical Science Building Rm 308A, 635 Barnhill Drive, Indianapolis, IN 46202-5120 USA
- Indiana University School of Medicine Center for Diabetes Research, Indianapolis, IN USA
| | - P. J. Gallagher
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, VanNuys Medical Science Building Rm 308A, 635 Barnhill Drive, Indianapolis, IN 46202-5120 USA
- Indiana University School of Medicine Center for Diabetes Research, Indianapolis, IN USA
| | - M. Sturek
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, VanNuys Medical Science Building Rm 308A, 635 Barnhill Drive, Indianapolis, IN 46202-5120 USA
- Indiana University School of Medicine Center for Diabetes Research, Indianapolis, IN USA
- Indiana University School of Medicine Center for Vascular Biology and Medicine, Indianapolis, IN USA
| | - M. A. Alloosh
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, VanNuys Medical Science Building Rm 308A, 635 Barnhill Drive, Indianapolis, IN 46202-5120 USA
- Indiana University School of Medicine Center for Diabetes Research, Indianapolis, IN USA
- Indiana University School of Medicine Center for Vascular Biology and Medicine, Indianapolis, IN USA
| | | | - R. V. Considine
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, VanNuys Medical Science Building Rm 308A, 635 Barnhill Drive, Indianapolis, IN 46202-5120 USA
- Indiana University School of Medicine Center for Diabetes Research, Indianapolis, IN USA
- Department of Medicine, Division of Endocrinology and Metabolism, Indiana University School of Medicine, Indianapolis, IN USA
| | - J. S. Elmendorf
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, VanNuys Medical Science Building Rm 308A, 635 Barnhill Drive, Indianapolis, IN 46202-5120 USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN USA
- Indiana University School of Medicine Center for Diabetes Research, Indianapolis, IN USA
- Indiana University School of Medicine Center for Membrane Biosciences, Indianapolis, IN USA
- Indiana University School of Medicine Center for Vascular Biology and Medicine, Indianapolis, IN USA
| |
Collapse
|
38
|
Kim JY, Kandror KV. The first luminal loop confers insulin responsiveness to glucose transporter 4. Mol Biol Cell 2012; 23:910-7. [PMID: 22262463 PMCID: PMC3290648 DOI: 10.1091/mbc.e11-10-0839] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Glucose transporter isoform 4 (GLUT4) is the sole glucose transporter responsible for the effect of insulin on postprandial blood glucose clearance. It is translocated to the plasma membrane by specialized insulin-responsive vesicles. Targeting of GLUT4 to these vesicles is mediated by sortilin, which interacts with the first luminal loop of the transporter. Glucose transporter isoform 4 (GLUT4), is the sole glucose transporter responsible for the effect of insulin on postprandial blood glucose clearance. However, the nature of the insulin sensitivity of GLUT4 remains unknown. In this study, we replaced the first luminal loop of cellugyrin, a 4-transmembrane protein that does not respond to insulin, with that of GLUT4. The chimera protein is targeted to the intracellular insulin-responsive vesicles and is translocated to the plasma membrane upon insulin stimulation. The faithful targeting of the chimera depends on the expression of the sorting receptor sortilin, which interacts with the unique amino acid residues in the first luminal loop of GLUT4. Thus the first luminal loop may confer insulin responsiveness to the GLUT4 molecule.
Collapse
Affiliation(s)
- Ju Youn Kim
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | | |
Collapse
|
39
|
Grant MP, Stepanchick A, Cavanaugh A, Breitwieser GE. Agonist-Driven Maturation and Plasma Membrane Insertion of Calcium-Sensing Receptors Dynamically Control Signal Amplitude. Sci Signal 2011; 4:ra78. [DOI: 10.1126/scisignal.2002208] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
40
|
Hatakeyama H, Kanzaki M. Molecular basis of insulin-responsive GLUT4 trafficking systems revealed by single molecule imaging. Traffic 2011; 12:1805-20. [PMID: 21910807 DOI: 10.1111/j.1600-0854.2011.01279.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Development of a 'static retention' property of GLUT4, the insulin-responsive glucose transporter, has emerged as being essential for achieving its maximal insulin-induced surface exposure. Herein, employing quantum-dot-based nanometrology of intracellular GLUT4 behavior, we reveal the molecular basis of its systematization endowed upon adipogenic differentiation of 3T3L1 cells. Specifically, (i) the endosomes-to-trans-Golgi network (TGN) retrieval system specialized for GLUT4 develops in response to sortilin expression, which requires an intricately balanced interplay among retromers, golgin-97 and syntaxin-6, the housekeeping vesicle trafficking machinery. (ii) The Golgin-97-localizing subdomain of the differentiated TGN apparently serves as an intermediate transit route by which GLUT4 can further proceed to the stationary GLUT4 storage compartment. (iii) AS160/Tbc1d4 then renders the 'static retention' property insulin responsive, i.e. insulin liberates GLUT4 from the static state only in the presence of functional AS160/Tbc1d4. (iv) Moreover, sortilin malfunction and the resulting GLUT4 sorting defects along with retarded TGN function might be etiologically related to insulin resistance. Together, these observations provide a conceptual framework for understanding maturation/retardation of the insulin-responsive GLUT4 trafficking system that relies on the specialized subdomain of differentiated TGN.
Collapse
Affiliation(s)
- Hiroyasu Hatakeyama
- Graduate School of Biomedical Engineering, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | | |
Collapse
|
41
|
Chiu TT, Jensen TE, Sylow L, Richter EA, Klip A. Rac1 signalling towards GLUT4/glucose uptake in skeletal muscle. Cell Signal 2011; 23:1546-54. [DOI: 10.1016/j.cellsig.2011.05.022] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Accepted: 05/31/2011] [Indexed: 12/27/2022]
|
42
|
Haga Y, Ishii K, Suzuki T. N-glycosylation is critical for the stability and intracellular trafficking of glucose transporter GLUT4. J Biol Chem 2011; 286:31320-7. [PMID: 21757715 DOI: 10.1074/jbc.m111.253955] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The facilitative glucose transporter GLUT4 plays a key role in regulating whole body glucose homeostasis. GLUT4 dramatically changes its distribution upon insulin stimulation, and insulin-resistant diabetes is often linked with compromised translocation of GLUT4 under insulin stimulation. To elucidate the functional significance of the sole N-glycan chain on GLUT4, wild-type GLUT4 and a GLUT4 glycosylation mutant conjugated with enhanced GFP were stably expressed in HeLa cells. The N-glycan contributed to the overall stability of newly synthesized GLUT4. Moreover, cell surface expression of wild-type GLUT4 in HeLa cells was elevated upon insulin treatment, whereas the glycosylation mutant lost the ability to respond to insulin. Subcellular distribution of the mutant was distinct from that of wild-type GLUT4, implying that the subcellular localization required for insulin-mediated translocation was impaired in the mutant protein. Interestingly, kifunensine-treated cells also lost sensitivity to insulin, suggesting the functional importance of the N-glycan structure for GLUT4 trafficking. The K(m) or turnover rates of wild-type and mutant GLUT4, however, were similar, suggesting that the N-glycan had little effect on transporter activity. These findings underscore the critical roles of the N-glycan chain in quality control as well as intracellular trafficking of GLUT4.
Collapse
Affiliation(s)
- Yoshimi Haga
- Glycometabolome Team, RIKEN Advanced Science Institute, Wako, Saitama 351-0198, Japan
| | | | | |
Collapse
|
43
|
Hyperinsulinemia leads to uncoupled insulin regulation of the GLUT4 glucose transporter and the FoxO1 transcription factor. Proc Natl Acad Sci U S A 2011; 108:10162-7. [PMID: 21646544 DOI: 10.1073/pnas.1019268108] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Insulin resistance is a component of the metabolic syndrome and Type 2 diabetes. It has been recently shown that in liver insulin resistance is not complete. This so-called selective insulin resistance is characterized by defective insulin inhibition of hepatic glucose output while insulin-induced lipogenesis is maintained. How this occurs and whether uncoupled insulin action develops in other tissues is unknown. Here we show in a model of chronic hyperinsulinemia that adipocytes develop selective insulin resistance in which translocation of the GLUT4 glucose transporter to the cell surface is blunted yet nuclear exclusion of the FoxO1 transcription factor is preserved, rendering uncoupled insulin-controlled carbohydrate and lipid metabolisms. We found that in adipocytes FoxO1 nuclear exclusion has a lower half-maximal insulin dose than GLUT4 translocation, and it is because of this inherent greater sensitivity that control of FoxO1 by physiological insulin concentrations is maintained in adipocytes with compromised insulin signaling. Pharmacological and genetic interventions revealed that insulin regulates GLUT4 and FoxO1 through the PI3-kinase isoform p110α, although FoxO1 showed higher sensitivity to p110α activity than GLUT4. Transient down-regulation and overexpression of Akt isoforms in adipocytes demonstrated that insulin-activated PI3-kinase signals to GLUT4 primarily through Akt2 kinase, whereas Akt1 and Akt2 signal to FoxO1. We propose that the lower threshold of insulin activity for FoxO1's nuclear exclusion is in part due to its regulation by both Akt isoforms. Identification of uncoupled insulin action in adipocytes suggests this condition might be a general phenomenon of insulin target tissues contributing to insulin resistance's pathophysiology.
Collapse
|
44
|
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
|
45
|
Abstract
Translocation of Glut4 to the plasma membrane of fat and skeletal muscle cells is mediated by specialized insulin-responsive vesicles (IRVs), whose protein composition consists primarily of glucose transporter isoform 4 (Glut4), insulin-responsive amino peptidase (IRAP), sortilin, lipoprotein receptor-related protein 1 (LRP1) and v-SNAREs. How can these proteins find each other in the cell and form functional vesicles after endocytosis from the plasma membrane? We are proposing a model according to which the IRV component proteins are internalized into sorting endosomes and are delivered to the IRV donor compartment(s), recycling endosomes and/or the trans-Golgi network (TGN), by cellugyrin-positive transport vesicles. The cytoplasmic tails of Glut4, IRAP, LRP1 and sortilin play an important targeting role in this process. Once these proteins arrive in the donor compartment, they interact with each other via their lumenal domains. This facilitates clustering of the IRV proteins into an oligomeric complex, which can then be distributed from the donor membranes to the IRV as a single entity with the help of adaptors, such as Golgi-localized, gamma-adaptin ear-containing, ARF-binding (GGA).
Collapse
Affiliation(s)
- Konstantin V Kandror
- Department of Biochemistry, Boston University School of Medicine, 72 E. Concord Street, Boston, MA 02118, USA
| | | |
Collapse
|
46
|
Hoffman NJ, Elmendorf JS. Signaling, cytoskeletal and membrane mechanisms regulating GLUT4 exocytosis. Trends Endocrinol Metab 2011; 22:110-6. [PMID: 21216617 PMCID: PMC3049829 DOI: 10.1016/j.tem.2010.12.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 12/03/2010] [Accepted: 12/06/2010] [Indexed: 10/25/2022]
Abstract
Solving how insulin regulates glucose transport into skeletal muscle and adipose tissue remains a fundamental challenge in biology and a significant issue in medicine. A central feature of this process is the coordinated accumulation of the glucose transporter GLUT4 into the plasma membrane. New signaling and cytoskeletal mechanisms of insulin-stimulated GLUT4 exocytosis are of emerging interest, particularly those at or just beneath the plasma membrane. This review examines signals that functionally engage GLUT4 exocytosis, considers cytoskeletal regulation of the stimulated GLUT4 itinerary, and appraises the involvement of plasma membrane parameters in GLUT4 control. We also explore how these newly-defined signaling, cytoskeletal and membrane mechanisms could be of therapeutic interest in the treatment and/or prevention of GLUT4 dysregulation in disease.
Collapse
Affiliation(s)
- Nolan J Hoffman
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Centers for Diabetes Research, Membrane Biosciences, and Vascular Biology and Medicine, VanNuys Medical Science Building Room 308A, Indianapolis, IN 46202, USA
| | | |
Collapse
|
47
|
Lin R, Murtazina R, Cha B, Chakraborty M, Sarker R, Chen TE, Lin Z, Hogema BM, de Jonge HR, Seidler U, Turner JR, Li X, Kovbasnjuk O, Donowitz M. D-glucose acts via sodium/glucose cotransporter 1 to increase NHE3 in mouse jejunal brush border by a Na+/H+ exchange regulatory factor 2-dependent process. Gastroenterology 2011; 140:560-71. [PMID: 20977906 PMCID: PMC3031713 DOI: 10.1053/j.gastro.2010.10.042] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Revised: 09/13/2010] [Accepted: 10/15/2010] [Indexed: 01/13/2023]
Abstract
BACKGROUND & AIMS Oral rehydration solutions reduce diarrhea-associated mortality. Stimulated sodium absorption by these solutions is mediated by the Na(+)/H(+) hydrogen exchanger NHE3 and is increased by Na(+)-glucose co-transport in vitro, but the mechanisms of this up-regulated process are only partially understood. METHODS Intracellular pH was measured in jejunal enterocytes of wild-type mice and mice with disrupted Na+/H+ exchange regulatory co-factor 2 (NHERF2-/- mice) by multiphoton microscopy. Diarrhea was induced by cholera toxin. Caco-2BBe cells that express NHE3 and the sodium/glucose cotransporter 1 (SGLT1) were studied by fluorometry, before and after siRNA-mediated knockdown of NHERF1 or NHERF2. NHE3 distribution was assessed by cell-surface biotinylation and confocal microscopy. Brush-border mobility was determined by fluorescence recovery after photobleaching and confocal microscopy. RESULTS The nonmetabolized SGLT1 substrate α-methyl-D-Glu (α-MD-G) activated jejunal NHE3; this process required Akt and NHERF2. α-MD-G normalized NHE3 activity after cholera toxin-induced diarrhea. α-MD-G-stimulated jejunal NHE3 activity was defective in NHERF2-/- mice and cells with NHERF2 knockdown, but occurred normally with NHERF1 knockdown; was associated with increased NHE3 surface expression in Caco-2 cells, which also was NHERF2-dependent; was associated with dissociation of NHE3 from NHERF2 and an increase in the NHE3 mobile fraction from the brush border; and was accompanied by a NHERF2 ezrin-radixin-moesin-binding domain-dependent increase in co-precipitation of ezrin with NHE3. CONCLUSIONS SGLT1-mediated Na-glucose co-transport stimulates NHE3 activity in vivo by an Akt- and NHERF2-dependent signaling pathway. It is associated with increased brush-border NHE3 and association between ezrin and NHE3. Activation of NHE3 corrects cholera toxin-induced defects in Na absorption and might contribute to the efficacy of oral rehydration solutions.
Collapse
Affiliation(s)
- Rong Lin
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA, GI Division, Wuhan Union Hospital, Tongi Medical School, Huazhong University of Science and Technology, 1277 Jiefang Road, Wuhan, China
| | - Rakhilya Murtazina
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Boyoung Cha
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Molee Chakraborty
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rafiquel Sarker
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tian-e Chen
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zhihong Lin
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Boris M. Hogema
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Hugo R. de Jonge
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Ursula Seidler
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Jerrold R. Turner
- Department of Physiology, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xuhang Li
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Olga Kovbasnjuk
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mark Donowitz
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA, GI Division, Wuhan Union Hospital, Tongi Medical School, Huazhong University of Science and Technology, 1277 Jiefang Road, Wuhan, China
| |
Collapse
|
48
|
Jordens I, Molle D, Xiong W, Keller SR, McGraw TE. Insulin-regulated aminopeptidase is a key regulator of GLUT4 trafficking by controlling the sorting of GLUT4 from endosomes to specialized insulin-regulated vesicles. Mol Biol Cell 2010; 21:2034-44. [PMID: 20410133 PMCID: PMC2883947 DOI: 10.1091/mbc.e10-02-0158] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
IRAP is a key regulator of GLUT4 trafficking by controlling sorting from endosomes to specialized insulin-regulated vesicles. Insulin stimulates glucose uptake by regulating translocation of the GLUT4 glucose transporter from intracellular compartments to the plasma membrane. In the absence of insulin GLUT4 is actively sequestered away from the general endosomes into GLUT4-specialized compartments, thereby controlling the amount of GLUT4 at the plasma membrane. Here, we investigated the role of the aminopeptidase IRAP in GLUT4 trafficking. In unstimulated IRAP knockdown adipocytes, plasma membrane GLUT4 levels are elevated because of increased exocytosis, demonstrating an essential role of IRAP in GLUT4 retention. Current evidence supports the model that AS160 RabGAP, which is required for basal GLUT4 retention, is recruited to GLUT4 compartments via an interaction with IRAP. However, here we show that AS160 recruitment to GLUT4 compartments and AS160 regulation of GLUT4 trafficking were unaffected by IRAP knockdown. These results demonstrate that AS160 is recruited to membranes by an IRAP-independent mechanism. Consistent with a role independent of AS160, we showed that IRAP functions in GLUT4 sorting from endosomes to GLUT4-specialized compartments. This is revealed by the relocalization of GLUT4 to endosomes in IRAP knockdown cells. Although IRAP knockdown has profound effects on GLUT4 traffic, GLUT4 knockdown does not affect IRAP trafficking, demonstrating that IRAP traffics independent of GLUT4. In sum, we show that IRAP is both cargo and a key regulator of the insulin-regulated pathway.
Collapse
Affiliation(s)
- Ingrid Jordens
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10065, USA
| | | | | | | | | |
Collapse
|
49
|
Biogenesis and regulation of insulin-responsive vesicles containing GLUT4. Curr Opin Cell Biol 2010; 22:506-12. [PMID: 20417083 DOI: 10.1016/j.ceb.2010.03.012] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Revised: 03/29/2010] [Accepted: 03/31/2010] [Indexed: 12/16/2022]
Abstract
Insulin regulates the trafficking of GLUT4 glucose transporters in fat and muscle cells. In unstimulated cells, GLUT4 is sequestered intracellularly in small, insulin-responsive vesicles. Insulin stimulates the translocation of these vesicles to the cell surface, inserting the transporters into the plasma membrane to enhance glucose uptake. Formation of the insulin-responsive vesicles requires multiple interactions among GLUT4, IRAP, LRP1, and sortilin, as well as recruitment of GGA and ACAP1 adaptors and clathrin. Once formed, the vesicles are retained within unstimulated cells by the action of TUG, Ubc9, and other proteins. In addition to acting at other steps in vesicle recycling, insulin releases this retention mechanism to promote the translocation and fusion of the vesicles at the cell surface.
Collapse
|