1
|
Xu Y, Yang T, Xu Q, Tang Y, Yang Q. Vesicle-associated membrane protein 8 knockdown exerts anti-proliferative, pro-apoptotic, anti-autophagic, and pro-ferroptotic effects on colorectal cancer cells by inhibition of the JAK/STAT3 pathway. J Bioenerg Biomembr 2024; 56:419-431. [PMID: 38720136 DOI: 10.1007/s10863-024-10019-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 04/20/2024] [Indexed: 07/03/2024]
Abstract
Vesicle-associated membrane protein 8 (VAMP8), a soluble n-ethylmaleimide-sensitive factor receptor protein, acts as an oncogenic gene in the progression of several malignancies. Nevertheless, the roles and mechanisms of VAMP8 in colorectal cancer (CRC) progression remain unknown. The expression and prognostic significance of VAMP8 in CRC samples were analyzed through bioinformatics analyses. Cell proliferation was detected using CCK-8 and EdU incorporation assays and apoptosis was evaluated via flow cytometry. Western blot analysis was conducted to examine the protein expression. Ferroptosis was evaluated by measurement of iron metabolism, lipid peroxidation, and glutathione (GSH) content. VAMP8 was increased in CRC samples relative to normal samples on the basis of GEPIA and HPA databases. CRC patients with high level of VAMP8 had a worse overall survival. VAMP8 depletion led to a suppression of proliferation and promotion of apoptosis in CRC cells. Additionally, VAMP8 knockdown suppressed beclin1 expression and LC3-II/LC3-I ratio, elevated p62 expression, increased Fe2+, labile iron pool, lipid reactive oxygen species, and malondialdehyde levels, and repressed GSH content and glutathione peroxidase activity. Moreover, VAMP8 knockdown inhibited the activation of janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) pathway in CRC cells. Mechanistically, activation of the JAK/STAT3 pathway by JAK1 or JAK2 overexpression attenuated VAMP8 silencing-mediated anti-proliferative, pro-apoptotic, anti-autophagic, and pro-ferroptotic effects on CRC cells. In conclusion, VAMP8 knockdown affects the proliferation, apoptosis, autophagy, and ferroptosis by the JAK/STAT3 pathway in CRC cells.
Collapse
Affiliation(s)
- Yi Xu
- Department of General Surgery, Nanyang First People's Hospital, Nanyang, China
| | - Tianyao Yang
- Department of General Surgery, Tiantai People's Hospital of Zhejiang Province, Taizhou, China
| | - Qiu Xu
- Department of Thyroid and Breast Surgery, Nanyang First People's Hospital, Nanyang, China
- Nanyang Key Laboratory of Thyroid Tumor Prevention and Treatment, Nanyang First People's Hospital, Nanyang, China
| | - Yan Tang
- Department of General Surgery, Nanyang First People's Hospital, Nanyang, China
| | - Qiong Yang
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China.
| |
Collapse
|
2
|
Joshi S, Prakhya KS, Smith AN, Chanzu H, Zhang M, Whiteheart SW. The complementary roles of VAMP-2, -3, and -7 in platelet secretion and function. Platelets 2023; 34:2237114. [PMID: 37545110 PMCID: PMC10564522 DOI: 10.1080/09537104.2023.2237114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/21/2023] [Accepted: 07/10/2023] [Indexed: 08/08/2023]
Abstract
Platelet secretion requires Soluble N-ethylmaleimide Sensitive Attachment Protein Receptors (SNAREs). Vesicle SNAREs/Vesicle-Associated Membrane Proteins (v-SNAREs/VAMPs) on granules and t-SNAREs in plasma membranes mediate granule release. Platelet VAMP heterogeneity has complicated the assessment of how/if each is used and affects hemostasis. To address the importance of VAMP-7 (V7), we analyzed mice with global deletions of V3 and V7 together or platelet-specific deletions of V2, V3, and global deletion of V7. We measured the kinetics of cargo release, and its effects on three injury models to define the context-specific roles of these VAMPs. Loss of V7 minimally affected dense and α granule release but did affect lysosomal release. V3-/-7-/- and V2Δ3Δ7-/- platelets showed partial defects in α and lysosomal release; dense granule secretion was unaffected. In vivo assays showed that loss of V2, V3, and V7 caused no bleeding or occlusive thrombosis. These data indicate a role for V7 in lysosome release that is partially compensated by V3. V7 and V3, together, contribute to α granule release, however none of these deletions affected hemostasis/thrombosis. Our results confirm the dominance of V8. When it is present, deletion of V2, V3, or V7 alone or in combination minimally affects platelet secretion and hemostasis.
Collapse
Affiliation(s)
- Smita Joshi
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | | | - Alexis N. Smith
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Harry Chanzu
- GenScript USA Inc., 860 Centennial Ave. Piscataway, NJ 08854, USA
| | - Ming Zhang
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Sidney W. Whiteheart
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
3
|
Bremner SK, Al Shammari WS, Milligan RS, Hudson BD, Sutherland C, Bryant NJ, Gould GW. Pleiotropic effects of Syntaxin16 identified by gene editing in cultured adipocytes. Front Cell Dev Biol 2022; 10:1033501. [PMID: 36467416 PMCID: PMC9716095 DOI: 10.3389/fcell.2022.1033501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/02/2022] [Indexed: 08/01/2023] Open
Abstract
Adipocytes play multiple roles in the regulation of glucose metabolism which rely on the regulation of membrane traffic. These include secretion of adipokines and serving as an energy store. Central to their energy storing function is the ability to increase glucose uptake in response to insulin, mediated through translocation of the facilitative glucose transporter GLUT4 to the cell surface. The trans-Golgi reticulum localized SNARE protein syntaxin 16 (Sx16) has been identified as a key component of the secretory pathway required for insulin-regulated trafficking of GLUT4. We used CRISPR/Cas9 technology to generate 3T3-L1 adipocytes lacking Sx16 to understand the role of the secretory pathway on adipocyte function. GLUT4 mRNA and protein levels were reduced in Sx16 knockout adipocytes and insulin stimulated GLUT4 translocation to the cell surface was reduced. Strikingly, neither basal nor insulin-stimulated glucose transport were affected. By contrast, GLUT1 levels were upregulated in Sx16 knockout cells. Levels of sortilin and insulin regulated aminopeptidase were also increased in Sx16 knockout adipocytes which may indicate an upregulation of an alternative GLUT4 sorting pathway as a compensatory mechanism for the loss of Sx16. In response to chronic insulin stimulation, Sx16 knockout adipocytes exhibit elevated insulin-independent glucose transport and significant alterations in lactate metabolism. We further show that the adipokine secretory pathways are impaired in Sx16 knockout cells. Together this demonstrates a role for Sx16 in the control of glucose transport, the response to elevated insulin, cellular metabolic profiles and adipocytokine secretion.
Collapse
Affiliation(s)
- Shaun K. Bremner
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Woroud S. Al Shammari
- The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow, United Kingdom
| | - Roderick S. Milligan
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Brian D. Hudson
- The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow, United Kingdom
| | - Calum Sutherland
- Department of Cellular Medicine, Ninewells Hospital, University of Dundee, Glasgow, United Kingdom
| | - Nia J. Bryant
- Department of Biology, University of York, York, United Kingdom
| | - Gwyn W. Gould
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
4
|
Liu F, He R, Zhu M, Zhou L, Liu Y, Yu H. Assembly-promoting protein Munc18c stimulates SNARE-dependent membrane fusion through its SNARE-like peptide. J Biol Chem 2022; 298:102470. [PMID: 36087838 PMCID: PMC9547204 DOI: 10.1016/j.jbc.2022.102470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 11/19/2022] Open
Abstract
Intracellular vesicle fusion requires the soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) and their cognate Sec1/Munc18 (SM) proteins. How SM proteins act in concert with trans-SNARE complexes to promote membrane fusion remains incompletely understood. Munc18c, a broadly distributed SM protein, selectively regulates multiple exocytotic pathways, including GLUT4 exocytosis. Here, using an in vitro reconstituted system, we discovered a SNARE-like peptide (SLP), conserved in Munc18-1 of synaptic exocytosis, is crucial to the stimulatory activity of Munc18c in vesicle fusion. The direct stimulation of the SNARE-mediated fusion reaction by SLP further supported the essential role of this fragment. Interestingly, we found SLP strongly accelerates the membrane fusion rate when anchored to the target membrane but not the vesicle membrane, suggesting it primarily interacts with t-SNAREs in cis to drive fusion. Furthermore, we determined the SLP fragment is competitive with the full-length Munc18c protein and specific to the cognate v-SNARE isoforms, supporting how it could resemble Munc18c’s activity in membrane fusion. Together, our findings demonstrate that Munc18c facilitates SNARE-dependent membrane fusion through SLP, revealing that the t-SNARE-SLP binding mode might be a conserved mechanism for the stimulatory function of SM proteins in vesicle fusion.
Collapse
Affiliation(s)
- Furong Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Ruyue He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Min Zhu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Lin Zhou
- School of Chemistry and Bioengineering, Nanjing Normal University Taizhou College, Taizhou, China
| | - Yinghui Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China.
| | - Haijia Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China.
| |
Collapse
|
5
|
Three live-imaging techniques for comprehensively understanding the initial trigger for insulin-responsive intracellular GLUT4 trafficking. iScience 2022; 25:104164. [PMID: 35434546 PMCID: PMC9010770 DOI: 10.1016/j.isci.2022.104164] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 11/16/2021] [Accepted: 03/24/2022] [Indexed: 01/31/2023] Open
Abstract
Quantitative features of GLUT4 glucose transporter's behavior deep inside cells remain largely unknown. Our previous analyses with live-cell imaging of intracellular GLUT4 trafficking demonstrated two crucial early events responsible for triggering insulin-responsive translocation processes, namely, heterotypic fusion and liberation. To quantify the regulation, interrelationships, and dynamics of the initial events more accurately and comprehensively, we herein applied three analyses, each based on our distinct dual-color live-cell imaging approaches. With these approaches, heterotypic fusion was found to be the first trigger for insulin-responsive GLUT4 redistributions, preceding liberation, and to be critically regulated by Akt substrate of 160 kDa (AS160) and actin dynamics. In addition, demonstrating the subcellular regional dependence of GLUT4 dynamics revealed that liberated GLUT4 molecules are promptly incorporated into the trafficking itinerary of transferrin receptors. Our approaches highlight the physiological significance of endosomal "GLUT4 molecule trafficking" rather than "GLUT4 vesicle delivery" to the plasma membrane in response to insulin.
Collapse
|
6
|
Liu Y, He R, Zhu M, Yu H. In Vitro Reconstitution Studies of SNAREs and Their Regulators Mediating GLUT4 Vesicle Fusion. Methods Mol Biol 2022; 2473:141-156. [PMID: 35819764 DOI: 10.1007/978-1-0716-2209-4_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The GLUT4 vesicle fusion is mediated by soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) and a variety of regulatory proteins. For example, synip and tomosyn negatively regulate GLUT4 SNARE-mediated membrane fusion. Here we describe in vitro reconstituted assays to determine the molecular mechanisms of SNAREs, synip, and tomosyn. These methods can also be extended to the studies of other types of membrane fusion events.
Collapse
Affiliation(s)
- Yinghui Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Ruyue He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Min Zhu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Haijia Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China.
| |
Collapse
|
7
|
Black HL, Livingstone R, Mastick CC, Al Tobi M, Taylor H, Geiser A, Stirrat L, Kioumourtzoglou D, Petrie JR, Boyle JG, Bryant NJ, Gould GW. Knockout of Syntaxin-4 in 3T3-L1 adipocytes reveals new insight into GLUT4 trafficking and adiponectin secretion. J Cell Sci 2021; 135:273617. [PMID: 34859814 PMCID: PMC8767277 DOI: 10.1242/jcs.258375] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 11/18/2021] [Indexed: 11/20/2022] Open
Abstract
Adipocytes are key to metabolic regulation, exhibiting insulin-stimulated glucose transport that is underpinned by the insulin-stimulated delivery of glucose transporter type 4 (SLC2A4, also known and hereafter referred to as GLUT4)-containing vesicles to the plasma membrane where they dock and fuse, and increase cell surface GLUT4 levels. Adipocytokines, such as adiponectin, are secreted via a similar mechanism. We used genome editing to knock out syntaxin-4, a protein reported to mediate fusion between GLUT4-containing vesicles and the plasma membrane in 3T3-L1 adipocytes. Syntaxin-4 knockout reduced insulin-stimulated glucose transport and adiponectin secretion by ∼50% and reduced GLUT4 levels. Ectopic expression of haemagglutinin (HA)-tagged GLUT4 conjugated to GFP showed that syntaxin-4-knockout cells retain significant GLUT4 translocation capacity, demonstrating that syntaxin-4 is dispensable for insulin-stimulated GLUT4 translocation. Analysis of recycling kinetics revealed only a modest reduction in the exocytic rate of GLUT4 in knockout cells, and little effect on endocytosis. These analyses demonstrate that syntaxin-4 is not always rate limiting for GLUT4 delivery to the cell surface. In sum, we show that syntaxin-4 knockout results in reduced insulin-stimulated glucose transport, depletion of cellular GLUT4 levels and inhibition of adiponectin secretion but has only modest effects on the translocation capacity of the cells. This article has an associated First Person interview with Hannah L. Black and Rachel Livingstone, joint first authors of the paper. Summary: Syntaxin-4 knockout reduces insulin-stimulated glucose transport, depletes levels of cellular GLUT4 and inhibits secretion of adiponectin but only modestly affects the translocation capacity of the cells.
Collapse
Affiliation(s)
- Hannah L Black
- Department of Biology and York Biomedical Research Institute, University of York. Heslington, York, YO10 5DD, UK
| | - Rachel Livingstone
- Henry Welcome Laboratory for Cell Biology, Institute for Molecular, Cellular and Systems Biology, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Cynthia C Mastick
- Henry Welcome Laboratory for Cell Biology, Institute for Molecular, Cellular and Systems Biology, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK.,Department of Biology, University of Nevada Reno, 1664 N. Virginia Street, Reno, NV 89557, USA
| | - Mohammed Al Tobi
- Henry Welcome Laboratory for Cell Biology, Institute for Molecular, Cellular and Systems Biology, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Holly Taylor
- Strathclyde Institute for Pharmacy and Biomedical Sciences, 161 Cathedral Street, University of Strathclyde, Glasgow G4 0RE, UK
| | - Angéline Geiser
- Strathclyde Institute for Pharmacy and Biomedical Sciences, 161 Cathedral Street, University of Strathclyde, Glasgow G4 0RE, UK
| | - Laura Stirrat
- Strathclyde Institute for Pharmacy and Biomedical Sciences, 161 Cathedral Street, University of Strathclyde, Glasgow G4 0RE, UK
| | - Dimitrios Kioumourtzoglou
- Department of Biology and York Biomedical Research Institute, University of York. Heslington, York, YO10 5DD, UK
| | - John R Petrie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow. Glasgow G12 8QQ, UK
| | - James G Boyle
- Institute of Cardiovascular and Medical Sciences, University of Glasgow. Glasgow G12 8QQ, UK.,School of Medicine, Dentistry and Nursing, University of Glasgow. Glasgow G12 8QQ, UK
| | - Nia J Bryant
- Department of Biology and York Biomedical Research Institute, University of York. Heslington, York, YO10 5DD, UK
| | - Gwyn W Gould
- Strathclyde Institute for Pharmacy and Biomedical Sciences, 161 Cathedral Street, University of Strathclyde, Glasgow G4 0RE, UK
| |
Collapse
|
8
|
Wang S, Liu Y, Crisman L, Wan C, Miller J, Yu H, Shen J. Genetic evidence for an inhibitory role of tomosyn in insulin-stimulated GLUT4 exocytosis. Traffic 2021; 21:636-646. [PMID: 32851733 DOI: 10.1111/tra.12760] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/21/2020] [Accepted: 08/22/2020] [Indexed: 12/11/2022]
Abstract
Exocytosis is a vesicle fusion process driven by soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs). A classic exocytic pathway is insulin-stimulated translocation of the glucose transporter type 4 (GLUT4) from intracellular vesicles to the plasma membrane in adipocytes and skeletal muscles. The GLUT4 exocytic pathway plays a central role in maintaining blood glucose homeostasis and is compromised in insulin resistance and type 2 diabetes. A candidate regulator of GLUT4 exocytosis is tomosyn, a soluble protein expressed in adipocytes. Tomosyn directly binds to GLUT4 exocytic SNAREs in vitro but its role in GLUT4 exocytosis was unknown. In this work, we used CRISPR-Cas9 genome editing to delete the two tomosyn-encoding genes in adipocytes. We observed that both basal and insulin-stimulated GLUT4 exocytosis was markedly elevated in the double knockout (DKO) cells. By contrast, adipocyte differentiation and insulin signaling remained intact in the DKO adipocytes. In a reconstituted liposome fusion assay, tomosyn inhibited all the SNARE complexes underlying GLUT4 exocytosis. The inhibitory activity of tomosyn was relieved by NSF and α-SNAP, which act in concert to remove tomosyn from GLUT4 exocytic SNAREs. Together, these studies revealed an inhibitory role for tomosyn in insulin-stimulated GLUT4 exocytosis in adipocytes. We suggest that tomosyn-arrested SNAREs represent a reservoir of fusion capacity that could be harnessed to treat patients with insulin resistance and type 2 diabetes.
Collapse
Affiliation(s)
- Shifeng Wang
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA.,Department of Chinese Medicine Information Science, Beijing University of Chinese Medicine, Beijing, China
| | - Yinghui Liu
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Lauren Crisman
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Chun Wan
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Jessica Miller
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Haijia Yu
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Jingshi Shen
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| |
Collapse
|
9
|
Huang Y, Zhou T, Zhang Y, Huang H, Ma Y, Wu C, Wang Q, Lin Q, Yang X, Pang K. Antidiabetic activity of a Flavonoid-Rich extract from flowers of Wisteria sinensis in type 2 diabetic mice via activation of the IRS-1/PI3K/Akt/GLUT4 pathway. J Funct Foods 2021. [DOI: 10.1016/j.jff.2020.104338] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
10
|
Complexin-2 redistributes to the membrane of muscle cells in response to insulin and contributes to GLUT4 translocation. Biochem J 2021; 478:407-422. [DOI: 10.1042/bcj20200542] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/11/2020] [Accepted: 01/04/2021] [Indexed: 11/17/2022]
Abstract
Insulin stimulates glucose uptake in muscle cells by rapidly redistributing vesicles containing GLUT4 glucose transporters from intracellular compartments to the plasma membrane (PM). GLUT4 vesicle fusion requires the formation of SNARE complexes between vesicular VAMP and PM syntaxin4 and SNAP23. SNARE accessory proteins usually regulate vesicle fusion processes. Complexins aide in neuro-secretory vesicle-membrane fusion by stabilizing trans-SNARE complexes but their participation in GLUT4 vesicle fusion is unknown. We report that complexin-2 is expressed and homogeneously distributed in L6 rat skeletal muscle cells. Upon insulin stimulation, a cohort of complexin-2 redistributes to the PM. Complexin-2 knockdown markedly inhibited GLUT4 translocation without affecting proximal insulin signalling of Akt/PKB phosphorylation and actin fiber remodelling. Similarly, complexin-2 overexpression decreased maximal GLUT4 translocation suggesting that the concentration of complexin-2 is finely tuned to vesicle fusion. These findings reveal an insulin-dependent regulation of GLUT4 insertion into the PM involving complexin-2.
Collapse
|
11
|
Liu W, Deng S, Liu Y, Zhao P, Ouyang Y, Yang X. Naphthalenes A and B, two new naphthalene derivatives from the fungus Phellinus igniarius. Nat Prod Res 2020; 36:3353-3359. [PMID: 33331172 DOI: 10.1080/14786419.2020.1858410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Two new naphthalene derivatives, named naphthalene A (1) and naphthalene B (2), together with eight known compounds (3-10) were isolated from dichloromethane fraction of Phellinus igniarius. The structures of compounds were elucidated on the basis of spectroscopic analysis and absolute configurations were established based on electronic circular dichroism data. Compounds 9 and 10 exhibited moderate activity, increasing Glucose transporter 4 (GLUT-4) translocation by 1.62 and 1.87 folds, respectively.
Collapse
Affiliation(s)
- Wei Liu
- Key Lab of Natural Product Chemistry and Application at Universities of Education Department of Xinjiang Uygur Autonomous Region, Yili Normal University, Yining, China
| | - Shihao Deng
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Yushuang Liu
- University of Chinese Academy of Sciences, Beijing, China
| | - Ping Zhao
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Yan Ouyang
- Key Lab of Natural Product Chemistry and Application at Universities of Education Department of Xinjiang Uygur Autonomous Region, Yili Normal University, Yining, China
| | - Xinzhou Yang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| |
Collapse
|
12
|
Guo J, Liu X, Zhang T, Lin X, Hong Y, Yu J, Wu Q, Zhang F, Wu Q, Shang J, Lv X, Ou J, Zhou J, Pang R, Tang B, Liang S. Hepatocyte TMEM16A Deletion Retards NAFLD Progression by Ameliorating Hepatic Glucose Metabolic Disorder. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903657. [PMID: 32440483 PMCID: PMC7237841 DOI: 10.1002/advs.201903657] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/22/2020] [Accepted: 02/27/2020] [Indexed: 06/11/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent form of chronic liver disease, and the mechanisms underpinning its pathogenesis have not been completely established. Transmembrane member 16A (TMEM16A), a component of the Ca2+-activated chloride channel (CaCC), has recently been implicated in metabolic events. Herein, TMEM16A is shown to be responsible for CaCC activation in hepatocytes and is increased in liver tissues of mice and patients with NAFLD. Hepatocyte-specific ablation of TMEM16A in mice ameliorates high-fat diet-induced obesity, hepatic glucose metabolic disorder, steatosis, insulin resistance, and inflammation. In contrast, hepatocyte-specific TMEM16A transgenic mice exhibit the opposite phenotype. Mechanistically, hepatocyte TMEM16A interacts with vesicle-associated membrane protein 3 (VAMP3) to induce its degradation, suppressing the formation of the VAMP3/syntaxin 4 and VAMP3/synaptosome-associated protein 23 complexes. This leads to the impairment of hepatic glucose transporter 2 (GLUT2) translocation and glucose uptake. Notably, VAMP3 overexpression restrains the functions of hepatocyte TMEM16A in blocking GLUT2 translocation and promoting lipid deposition, insulin resistance, and inflammation. In contrast, VAMP3 knockdown reverses the beneficial effects of TMEM16A downregulation. This study demonstrates a role for TMEM16A in NAFLD and suggests that inhibition of hepatic TMEM16A or disruption of TMEM16A/VAMP3 interaction may provide a new potential therapeutic strategy for NAFLD.
Collapse
Affiliation(s)
- Jia‐Wei Guo
- Department of PharmacologyCardiac and Cerebral Vascular Research CenterZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhou510080China
| | - Xiu Liu
- Department of PharmacologyCardiac and Cerebral Vascular Research CenterZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhou510080China
| | - Ting‐Ting Zhang
- Department of PharmacologyCardiac and Cerebral Vascular Research CenterZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhou510080China
| | - Xiao‐Chun Lin
- Department of PharmacologyCardiac and Cerebral Vascular Research CenterZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhou510080China
| | - Yu Hong
- Department of PharmacologyCardiac and Cerebral Vascular Research CenterZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhou510080China
| | - Jie Yu
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080China
| | - Qin‐Yan Wu
- Department of GastroenterologyThe First People's Hospital of FoshanFoshan528000China
| | - Fei‐Ran Zhang
- Department of PharmacologyCardiac and Cerebral Vascular Research CenterZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhou510080China
| | - Qian‐Qian Wu
- Key Laboratory of Metabolic Cardiovascular Diseases Research of National Health CommissionNingxia Medical UniversityYinchuan750004China
| | - Jin‐Yan Shang
- Department of PharmacologyCardiac and Cerebral Vascular Research CenterZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhou510080China
| | - Xiao‐Fei Lv
- Department of PharmacologyCardiac and Cerebral Vascular Research CenterZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhou510080China
| | - Jing‐Song Ou
- Division of Cardiac SurgeryThe Key Laboratory of Assisted CirculationMinistry of HealthThe First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080China
- National‐Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular DiseasesThe First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080China
| | - Jia‐Guo Zhou
- Department of PharmacologyCardiac and Cerebral Vascular Research CenterZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhou510080China
- Program of Kidney and Cardiovascular DiseaseThe Fifth Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080China
- Department of CardiologySun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120China
- Guangdong Province Key Laboratory of Brain Function and DiseaseZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhou510080China
| | - Rui‐Ping Pang
- Guangdong Province Key Laboratory of Brain Function and DiseaseZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhou510080China
- Department of PhysiologyPain Research CenterZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhou510080China
| | - Bao‐Dong Tang
- Department of GastroenterologyThe First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080China
| | - Si‐Jia Liang
- Department of PharmacologyCardiac and Cerebral Vascular Research CenterZhongshan School of MedicineSun Yat‐Sen UniversityGuangzhou510080China
| |
Collapse
|
13
|
Matthewson J. Detail and generality in mechanistic explanation. STUDIES IN HISTORY AND PHILOSOPHY OF SCIENCE 2020; 80:28-36. [PMID: 32383670 DOI: 10.1016/j.shpsa.2018.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 11/05/2017] [Accepted: 06/01/2018] [Indexed: 06/11/2023]
Abstract
This article is about the role of abstraction in mechanistic explanations. Abstraction is widely recognised as a necessary concession to the practicalities of scientific work, but some mechanist philosophers argue that it is also a positive explanatory feature in its own right. I claim that in as much as these arguments are based on the idea that mechanistic explanation exhibits a trade-off between fine-grained detail and generality, they are unsuccessful. Detail and generality both appear to be important sources of explanatory power, but investigators do not need to make a choice between these desiderata, at least when an explanation incorporates further detail through the decomposition of the mechanism's parts.
Collapse
Affiliation(s)
- John Matthewson
- School of Humanities, Massey University Auckland, Private Bag 102904, North Shore, Auckland, 0745, New Zealand.
| |
Collapse
|
14
|
Luiken JJFP, Nabben M, Neumann D, Glatz JFC. Understanding the distinct subcellular trafficking of CD36 and GLUT4 during the development of myocardial insulin resistance. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165775. [PMID: 32209364 DOI: 10.1016/j.bbadis.2020.165775] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/12/2020] [Accepted: 03/17/2020] [Indexed: 01/06/2023]
Abstract
CD36 and GLUT4 are the main cardiac trans-sarcolemmal transporters for long-chain fatty acids and glucose, respectively. Together they secure the majority of cardiac energy demands. Moreover, these transporters each represent key governing kinetic steps in cardiac fatty acid and glucose fluxes, thereby offering major sites of regulation. The underlying mechanism of this regulation involves a perpetual vesicle-mediated trafficking (recycling) of both transporters between intracellular stores (endosomes) and the cell surface. In the healthy heart, CD36 and GLUT4 translocation to the cell surface is under short-term control of the same physiological stimuli, most notably increased contraction and insulin secretion. However, under chronic lipid overload, a condition that accompanies a Western lifestyle, CD36 and GLUT4 recycling are affected distinctly, with CD36 being expelled to the sarcolemma while GLUT4 is imprisoned within the endosomes. Moreover, the increased CD36 translocation towards the cell surface is a key early step, setting the heart on a route towards insulin resistance and subsequent contractile dysfunction. Therefore, the proteins making up the trafficking machinery of CD36 need to be identified with special focus to the differences with the protein composition of the GLUT4 trafficking machinery. These proteins that are uniquely dedicated to either CD36 or GLUT4 traffic may offer targets to rectify aberrant substrate uptake seen in the lipid-overloaded heart. Specifically, CD36-dedicated trafficking regulators should be inhibited, whereas such GLUT4-dedicated proteins would need to be activated. Recent advances in the identification of CD36-dedicated trafficking proteins have disclosed the involvement of vacuolar-type H+-ATPase and of specific vesicle-associated membrane proteins (VAMPs). In this review, we summarize these recent findings and sketch a roadmap of CD36 and GLUT4 trafficking compatible with experimental findings.
Collapse
Affiliation(s)
- Joost J F P Luiken
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands.
| | - Miranda Nabben
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands; Department of Clinical Genetics, Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Centre, 6211 LK Maastricht, the Netherlands
| | - Dietbert Neumann
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6211 LK Maastricht, the Netherlands
| | - Jan F C Glatz
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands; Department of Clinical Genetics, Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Centre, 6211 LK Maastricht, the Netherlands
| |
Collapse
|
15
|
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
|
16
|
Henriksen TI, Wigge LV, Nielsen J, Pedersen BK, Sandri M, Scheele C. Dysregulated autophagy in muscle precursor cells from humans with type 2 diabetes. Sci Rep 2019; 9:8169. [PMID: 31160616 PMCID: PMC6546785 DOI: 10.1038/s41598-019-44535-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 04/12/2019] [Indexed: 12/18/2022] Open
Abstract
Autophagy is active during cellular remodeling including muscle differentiation. Muscle differentiation is dysregulated in type 2 diabetes and we therefore hypothesize that muscle precursor cells from people with type 2 diabetes (T2DM) have a dysregulation of their autophagy leading to impaired myogenesis. Muscle precursor cells were isolated from people with T2DM or healthy controls and differentiated in vitro. Autophagy marker levels were assessed by immunoblotting. Differentially expressed autophagy-related genes between healthy and T2DM groups were identified based on a previously published RNA-sequencing data-set, which we verified by RT-qPCR. siRNA was used to assess the function of differentially expressed autophagy genes. Basal autophagy increases during human muscle differentiation, while T2DM muscle cells have reduced levels of autophagy marker ATG7 and show a blunted response to starvation. Moreover, we demonstrate that the 3 non-canonical autophagy genes DRAM1, VAMP8 and TP53INP1 as differentially expressed between healthy and T2DM groups during myoblast differentiation, and that T53INP1 knock-down alters expression of both pro-and anti-apoptotic genes. In vitro differentiated T2DM muscle cells show differential expression of autophagy-related genes. These genes do not regulate myogenic transcription factors but may rather be involved in p53-associated myoblast apoptosis during early myogenesis.
Collapse
Affiliation(s)
- T I Henriksen
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
- Novo Nordisk Foundation Center, Section for Basic Metabolic Research, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark.
| | - L V Wigge
- Department of Biology and Biological Engineering, Chalmers University of Technology, 41296, Gothenburg, Sweden
| | - J Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, 41296, Gothenburg, Sweden
| | - B K Pedersen
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - M Sandri
- Venetian Institute of Molecular Medicine, via Orus 2, 35129, Padova, Italy
| | - C Scheele
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center, Section for Basic Metabolic Research, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
17
|
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
|
18
|
Yuan M, Liao J, Luo J, Cui M, Jin F. Significance of Vesicle-Associated Membrane Protein 8 Expression in Predicting Survival in Breast Cancer. J Breast Cancer 2018; 21:399-405. [PMID: 30607161 PMCID: PMC6310720 DOI: 10.4048/jbc.2018.21.e57] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 09/21/2018] [Indexed: 12/23/2022] Open
Abstract
Purpose Vesicle-associated membrane protein 8 (VAMP8) is a soluble N-ethylmaleimide-sensitive factor receptor protein that participates in autophagy by directly regulating autophagosome membrane fusion and has been reported to be involved in tumor progression. Nevertheless, the expression and prognostic value of VAMP8 in breast cancer (BC) remain unknown. This study aimed to evaluate the clinical significance and biological function of VAMP8 in BC. Methods A total of 112 BC samples and 30 normal mammary gland samples were collected. The expression of VAMP8 was assessed in both BC tissues and normal mammary gland tissues via a two-step immunohistochemical detection method. Results The expression of VAMP8 in BC tissues was significantly higher than that in normal breast tissues. Furthermore, increased VAMP8 expression was significantly correlated with tumor size (p=0.007), lymph node metastasis (p=0.024) and recurrence (p=0.001). Patients with high VAMP8 expression had significantly lower cumulative recurrence-free survival and overall survival (p<0.001 for both) than patients with low VAMP8 expression. In multivariate logistic regression and Cox regression analyses, lymph node metastasis and VAMP8 expression were independent prognostic factors for BC. Conclusion VAMP8 is significantly upregulated in human BC tissues and can thus be a practical and potentially effective surrogate marker for survival in BC patients.
Collapse
Affiliation(s)
- Mengci Yuan
- Division of Breast Surgery, Department of Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jianhua Liao
- Department of General Surgery, Zhejiang Hospital, Hangzhou, China
| | - Ji Luo
- Division of Breast Surgery, Department of Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Mengyao Cui
- Division of Breast Surgery, Department of Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Feng Jin
- Division of Breast Surgery, Department of Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
19
|
Zhong H, Wang P, Song Y, Zhang X, Che L, Feng B, Lin Y, Xu S, Li J, Wu D, Wu Q, Fang Z. Mammary cell proliferation and catabolism of adipose tissues in nutrition-restricted lactating sows were associated with extracellular high glutamate levels. J Anim Sci Biotechnol 2018; 9:78. [PMID: 30410753 PMCID: PMC6217789 DOI: 10.1186/s40104-018-0293-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 09/19/2018] [Indexed: 11/10/2022] Open
Abstract
Background Persistent lactation, as the result of mammary cellular anabolism and secreting function, is dependent on substantial mobilization or catabolism of body reserves under nutritional deficiency. However, little is known about the biochemical mechanisms for nutrition-restricted lactating animals to simultaneously maintain the anabolism of mammary cells while catabolism of body reserves. In present study, lactating sows with restricted feed allowance (RFA) (n = 6), 24% feed restriction compared with the control (CON) group (n = 6), were used as the nutrition-restricted model. Microdialysis and mammary venous cannulas methods were used to monitor postprandial dynamic changes of metabolites in adipose and mammary tissues. Results At lactation d 28, the RFA group showed higher (P < 0.05) loss of body weight and backfat than the CON group. Compared with the CON group, the adipose tissue of the RFA group had higher (P < 0.05) extracellular glutamate and insulin levels, increased (P < 0.05) lipolysis related genes (HSL and ATGL) expression, and decreased (P < 0.05) glucose transport and metabolism related genes (VAMP8, PKLR and LDHB) expression. These results indicated that under nutritional restriction, reduced insulin-mediated glucose uptake and metabolism and increased lipolysis in adipose tissues was related to extracellular high glutamate concentration. As for mammary glands, compared with the CON group, the RFA group had up-regulated (P < 0.05) expression of Notch signaling ligand (DLL3) and receptors (NOTCH2 and NOTCH4), higher (P < 0.05) extracellular glutamate concentration, while expression of cell proliferation related genes and concentrations of most metabolites in mammary veins were not different (P > 0.05) between groups. Accordingly, piglet performance and milk yield did not differ (P > 0.05) between groups. It would appear that activation of Notch signaling and adequate supply of glutamate might assist mammogenesis. Conclusions Mammary cell proliferation and catabolism of adipose tissues in nutrition-restricted lactating sows were associated with extracellular high glutamate levels. Electronic supplementary material The online version of this article (10.1186/s40104-018-0293-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Heju Zhong
- 1Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130 China
| | - Peng Wang
- 1Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130 China
| | - Yumo Song
- 1Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130 China
| | - Xiaoling Zhang
- 1Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130 China
| | - Lianqiang Che
- 1Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130 China
| | - Bin Feng
- 1Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130 China
| | - Yan Lin
- 1Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130 China
| | - Shengyu Xu
- 1Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130 China
| | - Jian Li
- 1Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130 China
| | - De Wu
- 1Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130 China
| | - Qiaofeng Wu
- 2Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137 China
| | - Zhengfeng Fang
- 1Key Laboratory for Animal Disease Resistance Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130 China
| |
Collapse
|
20
|
Genome-wide association meta-analysis of coronary artery disease and periodontitis reveals a novel shared risk locus. Sci Rep 2018; 8:13678. [PMID: 30209331 PMCID: PMC6135769 DOI: 10.1038/s41598-018-31980-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 08/31/2018] [Indexed: 02/07/2023] Open
Abstract
Evidence for a shared genetic basis of association between coronary artery disease (CAD) and periodontitis (PD) exists. To explore the joint genetic basis, we performed a GWAS meta-analysis. In the discovery stage, we used a German aggressive periodontitis sample (AgP-Ger; 680 cases vs 3,973 controls) and the CARDIoGRAMplusC4D CAD meta-analysis dataset (60,801 cases vs 123,504 controls). Two SNPs at the known CAD risk loci ADAMTS7 (rs11634042) and VAMP8 (rs1561198) passed the pre-assigned selection criteria (PAgP-Ger < 0.05; PCAD < 5 × 10−8; concordant effect direction) and were replicated in an independent GWAS meta-analysis dataset of PD (4,415 cases vs 5,935 controls). SNP rs1561198 showed significant association (PD[Replication]: P = 0.008 OR = 1.09, 95% CI = [1.02–1.16]; PD [Discovery + Replication]: P = 0.0002, OR = 1.11, 95% CI = [1.05–1.17]). For the associated haplotype block, allele specific cis-effects on VAMP8 expression were reported. Our data adds to the shared genetic basis of CAD and PD and indicate that the observed association of the two disease conditions cannot be solely explained by shared environmental risk factors. We conclude that the molecular pathway shared by CAD and PD involves VAMP8 function, which has a role in membrane vesicular trafficking, and is manipulated by pathogens to corrupt host immune defense.
Collapse
|
21
|
Huang Y, Hao J, Tian D, Wen Y, Zhao P, Chen H, Lv Y, Yang X. Antidiabetic Activity of a Flavonoid-Rich Extract From Sophora davidii (Franch.) Skeels in KK-Ay Mice via Activation of AMP-Activated Protein Kinase. Front Pharmacol 2018; 9:760. [PMID: 30061831 PMCID: PMC6055046 DOI: 10.3389/fphar.2018.00760] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/22/2018] [Indexed: 12/19/2022] Open
Abstract
The present study was undertaken to investigate the hypoglycemic activity and potential mechanisms of action of a flavonoid-rich extract from Sophora davidii (Franch.) Skeels (SD-FRE) through in vitro and in vivo studies. Four main flavonoids of SD-FRE namely apigenin, maackiain, leachianone A and leachianone B were purified and identified. In vitro, SD-FRE significantly promoted the translocation and expression of glucose transporter 4 (GLUT4) in L6 cells, which was significantly inhibited by Compound C (AMPK inhibitor), but not by Wortmannin (PI3K inhibitor) or Gö6983 (PKC inhibitor). These results indicated that SD-FRE enhanced GLUT4 expression and translocation to the plasma membrane via the AMPK pathway and finally resulted in an increase of glucose uptake. In vivo, using a spontaneously type 2 diabetic model, KK-Ay mice received intragastric administration of SD-FRE for 4 weeks. As a consequence, SD-FRE significantly alleviated the hyperglycemia, glucose intolerance, insulin resistance and hyperlipidemia in these mice. Hepatic steatosis, islet hypertrophy and larger adipocyte size were observed in KK-Ay mice. However, these pathological changes were effectively relieved by SD-FRE treatment. SD-FRE promoted GLUT4 expression and activated AMPK phosphorylation in insulin target tissues (muscle, adipose tissue and liver) of KK-Ay mice, thus facilitating glucose utilization to ameliorate insulin resistance. Regulation of ACC phosphorylation and PPARγ were also involved in the antidiabetic effects of SD-FRE. Taken together, these findings indicated that SD-FRE has the potential to alleviate type 2 diabetes.
Collapse
Affiliation(s)
- Yun Huang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Ji Hao
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Di Tian
- School of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Yanzhang Wen
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Ping Zhao
- School of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Hao Chen
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China.,College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Yibin Lv
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Xinzhou Yang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| |
Collapse
|
22
|
Su Z, Deshpande V, James DE, Stöckli J. Tankyrase modulates insulin sensitivity in skeletal muscle cells by regulating the stability of GLUT4 vesicle proteins. J Biol Chem 2018; 293:8578-8587. [PMID: 29669812 DOI: 10.1074/jbc.ra117.001058] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 04/05/2018] [Indexed: 11/06/2022] Open
Abstract
Tankyrase 1 and 2, members of the poly(ADP-ribose) polymerase family, have previously been shown to play a role in insulin-mediated glucose uptake in adipocytes. However, their precise mechanism of action, and their role in insulin action in other cell types, such as myocytes, remains elusive. Treatment of differentiated L6 myotubes with the small molecule tankyrase inhibitor XAV939 resulted in insulin resistance as determined by impaired insulin-stimulated glucose uptake. Proteomic analysis of XAV939-treated myotubes identified down-regulation of several glucose transporter GLUT4 storage vesicle (GSV) proteins including RAB10, VAMP8, SORT1, and GLUT4. A similar effect was observed following knockdown of tankyrase 1 in L6 myotubes. Inhibition of the proteasome using MG132 rescued GSV protein levels as well as insulin-stimulated glucose uptake in XAV939-treated L6 myotubes. These studies reveal an important role for tankyrase in maintaining the stability of key GLUT4 regulatory proteins that in turn plays a role in regulating cellular insulin sensitivity.
Collapse
Affiliation(s)
- Zhiduan Su
- From the Charles Perkins Centre, School of Life and Environmental Sciences and
| | - Vinita Deshpande
- From the Charles Perkins Centre, School of Life and Environmental Sciences and
| | - David E James
- From the Charles Perkins Centre, School of Life and Environmental Sciences and .,the Sydney Medical School, University of Sydney, Sydney 2006, Australia
| | - Jacqueline Stöckli
- From the Charles Perkins Centre, School of Life and Environmental Sciences and
| |
Collapse
|
23
|
CK2 modulates adipocyte insulin-signaling and is up-regulated in human obesity. Sci Rep 2017; 7:17569. [PMID: 29242563 PMCID: PMC5730587 DOI: 10.1038/s41598-017-17809-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 12/01/2017] [Indexed: 12/13/2022] Open
Abstract
Insulin plays a major role in glucose metabolism and insulin-signaling defects are present in obesity and diabetes. CK2 is a pleiotropic protein kinase implicated in fundamental cellular pathways and abnormally elevated in tumors. Here we report that in human and murine adipocytes CK2-inhibition decreases the insulin-induced glucose-uptake by counteracting Akt-signaling and GLUT4-translocation to the plasma membrane. In mice CK2 acts on insulin-signaling in adipose tissue, liver and skeletal muscle and its acute inhibition impairs glucose tolerance. Notably, CK2 protein-level and activity are greatly up-regulated in white adipose tissue from ob/ob and db/db mice as well as from obese patients, regardless the severity of their insulin-resistance and the presence of pre-diabetes or overt type 2 diabetes. Weight loss obtained by both bariatric surgery or hypocaloric diet reverts CK2 hyper-activation to normal level. Our data suggest a central role of CK2 in insulin-sensitivity, glucose homeostasis and adipose tissue remodeling. CK2 up-regulation is identified as a hallmark of adipose tissue pathological expansion, suggesting a new potential therapeutic target for human obesity.
Collapse
|
24
|
Li Y, Zhao P, Chen Y, Fu Y, Shi K, Liu L, Liu H, Xiong M, Liu QH, Yang G, Xiao Y. Depsidone and xanthones from Garcinia xanthochymus with hypoglycemic activity and the mechanism of promoting glucose uptake in L6 myotubes. Bioorg Med Chem 2017; 25:6605-6613. [DOI: 10.1016/j.bmc.2017.10.043] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/26/2017] [Accepted: 10/30/2017] [Indexed: 01/12/2023]
|
25
|
SNARE phosphorylation: a control mechanism for insulin-stimulated glucose transport and other regulated exocytic events. Biochem Soc Trans 2017; 45:1271-1277. [PMID: 29101310 DOI: 10.1042/bst20170202] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/03/2017] [Accepted: 10/04/2017] [Indexed: 11/17/2022]
Abstract
Trafficking within eukaryotic cells is a complex and highly regulated process; events such as recycling of plasma membrane receptors, formation of multivesicular bodies, regulated release of hormones and delivery of proteins to membranes all require directionality and specificity. The underpinning processes, including cargo selection, membrane fusion, trafficking flow and timing, are controlled by a variety of molecular mechanisms and engage multiple families of lipids and proteins. Here, we will focus on control of trafficking processes via the action of the SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) family of proteins, in particular their regulation by phosphorylation. We will describe how these proteins are controlled in a range of regulated trafficking events, with particular emphasis on the insulin-stimulated delivery of glucose transporters to the surface of adipose and muscle cells. Here, we focus on a few examples of SNARE phosphorylation which exemplify distinct ways in which SNARE machinery phosphorylation may regulate membrane fusion.
Collapse
|
26
|
Hatakeyama H, Kanzaki M. Heterotypic endosomal fusion as an initial trigger for insulin-induced glucose transporter 4 (GLUT4) translocation in skeletal muscle. J Physiol 2017; 595:5603-5621. [PMID: 28556933 DOI: 10.1113/jp273985] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 05/26/2017] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Comprehensive imaging analyses of glucose transporter 4 (GLUT4) behaviour in mouse skeletal muscle was conducted. Quantum dot-based single molecule nanometry revealed that GLUT4 molecules in skeletal myofibres are governed by regulatory systems involving 'static retention' and 'stimulus-dependent liberation'. Vital imaging analyses and super-resolution microscopy-based morphometry demonstrated that insulin liberates the GLUT4 molecule from its static state by triggering acute heterotypic endomembrane fusion arising from the very small GLUT4-containing vesicles in skeletal myofibres. Prior exposure to exercise-mimetic stimuli potentiated this insulin-responsive endomembrane fusion event involving GLUT4-containing vesicles, suggesting that this endomembranous regulation process is a potential site related to the effects of exercise. ABSTRACT Skeletal muscle is the major systemic glucose disposal site. Both insulin and exercise facilitate translocation of the glucose transporter glucose transporter 4 (GLUT4) via distinct signalling pathways and exercise also enhances insulin sensitivity. However, the trafficking mechanisms controlling GLUT4 mobilization in skeletal muscle remain poorly understood as a resuly of technical limitations. In the present study, which employs various imaging techniques on isolated skeletal myofibres, we show that one of the initial triggers of insulin-induced GLUT4 translocation is heterotypic endomembrane fusion arising from very small static GLUT4-containing vesicles with a subset of transferrin receptor-containing endosomes. Importantly, pretreatment with exercise-mimetic stimuli potentiated the susceptibility to insulin responsiveness, as indicated by these acute endomembranous activities. We also found that AS160 exhibited stripe-like localization close to sarcomeric α-actinin and that insulin induced a reduction of the stripe-like localization accompanying changes in its detergent solubility. The results of the present study thus provide a conceptual framework indicating that GLUT4 protein trafficking via heterotypic fusion is a critical feature of GLUT4 translocation in skeletal muscles and also suggest that the efficacy of the endomembranous fusion process in response to insulin is involved in the benefits of exercise.
Collapse
Affiliation(s)
- Hiroyasu Hatakeyama
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Japan.,Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | - Makoto Kanzaki
- Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| |
Collapse
|
27
|
Beg M, Abdullah N, Thowfeik FS, Altorki NK, McGraw TE. Distinct Akt phosphorylation states are required for insulin regulated Glut4 and Glut1-mediated glucose uptake. eLife 2017; 6. [PMID: 28589878 PMCID: PMC5462539 DOI: 10.7554/elife.26896] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/22/2017] [Indexed: 01/01/2023] Open
Abstract
Insulin, downstream of Akt activation, promotes glucose uptake into fat and muscle cells to lower postprandial blood glucose, an enforced change in cellular metabolism to maintain glucose homeostasis. This effect is mediated by the Glut4 glucose transporter. Growth factors also enhance glucose uptake to fuel an anabolic metabolism required for tissue growth and repair. This activity is predominantly mediated by the Glut1. Akt is activated by phosphorylation of its kinase and hydrophobic motif (HM) domains. We show that insulin-stimulated Glut4-mediated glucose uptake requires PDPK1 phosphorylation of the kinase domain but not mTORC2 phosphorylation of the HM domain. Nonetheless, an intact HM domain is required for Glut4-mediated glucose uptake. Whereas, Glut1-mediated glucose uptake also requires mTORC2 phosphorylation of the HM domain, demonstrating both phosphorylation-dependent and independent roles of the HM domain in regulating glucose uptake. Thus, mTORC2 links Akt to the distinct physiologic programs related to Glut4 and Glut1-mediated glucose uptake. DOI:http://dx.doi.org/10.7554/eLife.26896.001
Collapse
Affiliation(s)
- Muheeb Beg
- Department of Biochemistry, Weill Cornell Medicine, New York, United States
| | - Nazish Abdullah
- Department of Biochemistry, Weill Cornell Medicine, New York, United States
| | - Fathima Shazna Thowfeik
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, United States.,Lung Cancer Program, Meyer Cancer Center, Weill Cornell Medicine, New York, United States
| | - Nasser K Altorki
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, United States.,Lung Cancer Program, Meyer Cancer Center, Weill Cornell Medicine, New York, United States
| | - Timothy E McGraw
- Department of Biochemistry, Weill Cornell Medicine, New York, United States.,Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, United States.,Lung Cancer Program, Meyer Cancer Center, Weill Cornell Medicine, New York, United States
| |
Collapse
|
28
|
Zhou X, Shentu P, Xu Y. Spatiotemporal Regulators for Insulin-Stimulated GLUT4 Vesicle Exocytosis. J Diabetes Res 2017; 2017:1683678. [PMID: 28529958 PMCID: PMC5424486 DOI: 10.1155/2017/1683678] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/21/2017] [Accepted: 04/03/2017] [Indexed: 11/30/2022] Open
Abstract
Insulin increases glucose uptake and storage in muscle and adipose cells, which is accomplished through the mobilization of intracellular GLUT4 storage vesicles (GSVs) to the cell surface upon stimulation. Importantly, the dysfunction of insulin-regulated GLUT4 trafficking is strongly linked with peripheral insulin resistance and type 2 diabetes in human. The insulin signaling pathway, key signaling molecules involved, and precise trafficking itinerary of GSVs are largely identified. Understanding the interaction between insulin signaling molecules and key regulatory proteins that are involved in spatiotemporal regulation of GLUT4 vesicle exocytosis is of great importance to explain the pathogenesis of diabetes and may provide new potential therapeutic targets.
Collapse
Affiliation(s)
- Xiaoxu Zhou
- Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Ping Shentu
- Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Yingke Xu
- Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
- *Yingke Xu:
| |
Collapse
|
29
|
VAMP2 is implicated in the secretion of antibodies by human plasma cells and can be replaced by other synaptobrevins. Cell Mol Immunol 2016; 15:353-366. [PMID: 27616736 DOI: 10.1038/cmi.2016.46] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 07/06/2016] [Accepted: 07/06/2016] [Indexed: 11/08/2022] Open
Abstract
The production and secretion of antibodies by human plasma cells (PCs) are two essential processes of humoral immunity. The secretion process relies on a group of proteins known as soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs), which are located in the plasma membrane (t-SNAREs) and in the antibody-carrying vesicle membrane (v-SNARE), and mediate the fusion of both membranes. We have previously shown that SNAP23 and STX4 are the t-SNAREs responsible for antibody secretion. Here, using human PCs and antibody-secreting cell lines, we studied and characterized the expression and subcellular distribution of vesicle associated membrane protein (VAMP) isoforms, demonstrating that all isoforms (with the exception of VAMP1) are expressed by the referenced cells. Furthermore, the functional role in antibody secretion of each expressed VAMP isoform was tested using siRNA. Our results show that VAMP2 may be the v-SNARE involved in vesicular antibody release. To further support this conclusion, we used tetanus toxin light chain to cleave VAMP2, conducted experiments to verify co-localization of VAMP2 in antibody-carrying vesicles, and demonstrated the coimmunoprecipitation of VAMP2 with STX4 and SNAP23 and the in situ interaction of VAMP2 with STX4. Taken together, these findings implicate VAMP2 as the main VAMP isoform functionally involved in antibody secretion.
Collapse
|
30
|
Caceres PS, Mendez M, Haque MZ, Ortiz PA. Vesicle-associated Membrane Protein 3 (VAMP3) Mediates Constitutive Trafficking of the Renal Co-transporter NKCC2 in Thick Ascending Limbs: ROLE IN RENAL FUNCTION AND BLOOD PRESSURE. J Biol Chem 2016; 291:22063-22073. [PMID: 27551042 PMCID: PMC5063989 DOI: 10.1074/jbc.m116.735167] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Indexed: 02/04/2023] Open
Abstract
Renal cells of the thick ascending limb (TAL) reabsorb NaCl via the apical Na+/K+/2Cl- co-transporter NKCC2. Trafficking of NKCC2 to the apical surface regulates NKCC2-mediated NaCl absorption and blood pressure. The molecular mechanisms by which NKCC2 reaches the apical surface and their role in renal function and maintenance of blood pressure are poorly characterized. Here we report that NKCC2 interacts with the vesicle fusion protein VAMP3, and they co-localize at the TAL apical surface. We observed that silencing VAMP3 in vivo blocks constitutive NKCC2 exocytic delivery, decreasing the amount of NKCC2 at the TAL apical surface. VAMP3 is not required for cAMP-stimulated NKCC2 exocytic delivery. Additionally, genetic deletion of VAMP3 in mice decreased total expression of NKCC2 in the TAL and lowered blood pressure. Consistent with these results, urinary excretion of water and electrolytes was higher in VAMP3 knock-out mice, which produced more diluted urine. We conclude that VAMP3 interacts with NKCC2 and mediates its constitutive exocytic delivery to the apical surface. Additionally, VAMP3 is required for normal NKCC2 expression, renal function, and blood pressure.
Collapse
Affiliation(s)
- Paulo S Caceres
- From the Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan 48202, the Department of Physiology, Wayne State University, Detroit, Michigan 48202, and
| | - Mariela Mendez
- From the Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan 48202
| | - Mohammed Z Haque
- From the Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan 48202, the Interim Translational Research Institute, Academic Health System, Hamad Medical Corporation, 16060 Doha, Qatar
| | - Pablo A Ortiz
- From the Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan 48202, the Department of Physiology, Wayne State University, Detroit, Michigan 48202, and
| |
Collapse
|
31
|
Wilson JD, Shelby SA, Holowka D, Baird B. Rab11 Regulates the Mast Cell Exocytic Response. Traffic 2016; 17:1027-41. [PMID: 27288050 DOI: 10.1111/tra.12418] [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: 03/23/2016] [Revised: 06/07/2016] [Accepted: 06/07/2016] [Indexed: 01/30/2023]
Abstract
Stimulated exocytic events provide a means for physiological communication and are a hallmark of the mast cell-mediated allergic response. In mast cells these processes are triggered by antigen crosslinking of IgE bound to its high-affinity receptor, FcϵRI, on the cell surface. Here we use the endosomal v-SNARE VAMP8, and the lysosomal hydrolase β-hexosaminidase (β-Hex), each C-terminally fused to super-ecliptic pHluorin, to monitor stimulated exocytosis. Using these pHluorin-tagged constructs, we monitor stimulated exocytosis by fluorimetry and visualize individual exocytic events with total internal reflection (TIRF) microscopy. Similar to constitutive recycling endosome (RE) trafficking, we find that stimulated RE exocytosis, monitored by VAMP8, is attenuated by expression of dominant negative (S25N) Rab11. Stimulated β-Hex exocytosis is also reduced in the presence of S25N Rab11, suggesting that expression of this mutant broadly impacts exocytosis. Interestingly, pretreatment with inhibitors of actin polymerization, cytochalasin D or latrunculin A, substantially restores both RE and lysosome exocytosis in cells expressing S25N Rab11. Conversely, stabilizing F-actin with jasplakinolide inhibits antigen-stimulated exocytosis but is not additive with S25N Rab11-mediated inhibition, suggesting that these reagents inhibit related processes. Together, our results suggest that Rab11 participates in the regulation necessary for depolymerization of the actin cytoskeleton during stimulated exocytosis in mast cells.
Collapse
Affiliation(s)
- Joshua D Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853-1301, USA
| | - Sarah A Shelby
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853-1301, USA
| | - David Holowka
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853-1301, USA
| | - Barbara Baird
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853-1301, USA
| |
Collapse
|
32
|
Yang Z, Hong LK, Follett J, Wabitsch M, Hamilton NA, Collins BM, Bugarcic A, Teasdale RD. Functional characterization of retromer in GLUT4 storage vesicle formation and adipocyte differentiation. FASEB J 2015; 30:1037-50. [PMID: 26581601 DOI: 10.1096/fj.15-274704] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 10/28/2015] [Indexed: 12/26/2022]
Abstract
Insulin-stimulated translocation of glucose transporter 4 (GLUT4) storage vesicles (GSVs), the specialized intracellular compartments within mature adipocytes, to the plasma membrane (PM) is a fundamental cellular process for maintaining glucose homeostasis. Using 2 independent adipocyte cell line models, human primary Simpson-Golabi-Behmel syndrome and mouse 3T3-L1 fibroblast cell lines, we demonstrate that the endosome-associated protein-sorting complex retromer colocalizes with GLUT4 on the GSVs by confocal microscopy in mature adipocytes. By use of both confocal microscopy and differential ultracentrifugation techniques, retromer is redistributed to the PM of mature adipocytes upon insulin stimulation. Furthermore, stable knockdown of the retromer subunit-vacuolar protein-sorting 35, or the retromer-associated protein sorting nexin 27, by lentivirus-delivered small hairpin RNA impaired the adipogenesis process when compared to nonsilence control. The knockdown of retromer decreased peroxisome proliferator activated receptor γ expression during differentiation, generating adipocytes with decreased levels of GSVs, lipid droplet accumulation, and insulin-stimulated glucose uptake. In conclusion, our study demonstrates a role for retromer in the GSV formation and adipogenesis.
Collapse
Affiliation(s)
- Zhe Yang
- *Institute for Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia; and Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Lee Kian Hong
- *Institute for Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia; and Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Jordan Follett
- *Institute for Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia; and Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Martin Wabitsch
- *Institute for Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia; and Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Nicholas A Hamilton
- *Institute for Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia; and Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Brett M Collins
- *Institute for Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia; and Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Andrea Bugarcic
- *Institute for Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia; and Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Rohan D Teasdale
- *Institute for Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia; and Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| |
Collapse
|
33
|
Ropert N, Jalil A, Li D. Expression and cellular function of vSNARE proteins in brain astrocytes. Neuroscience 2015; 323:76-83. [PMID: 26518463 DOI: 10.1016/j.neuroscience.2015.10.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/07/2015] [Accepted: 10/21/2015] [Indexed: 12/27/2022]
Abstract
Gray matter protoplasmic astrocytes, a major type of glial cell in the mammalian brain, extend thin processes ensheathing neuronal synaptic terminals. Albeit electrically silent, astrocytes respond to neuronal activity with Ca(2+) signals that trigger the release of gliotransmitters, such as glutamate, d-serine, and ATP, which modulate synaptic transmission. It has been suggested that the astrocytic processes, together with neuronal pre- and post-synaptic elements, constitute a tripartite synapse, and that astrocytes actively regulate information processing. Astrocytic vesicles expressing VAMP2 and VAMP3 vesicular SNARE (vSNARE) proteins have been suggested to be a key feature of the tripartite synapse and mediate gliotransmitter release through Ca(2+)-regulated exocytosis. However, the concept of exocytotic release of gliotransmitters by astrocytes has been challenged. Here we review studies investigating the expression profile of VAMP2 and VAMP3 vSNARE proteins in rodent astrocytes, and the functional implication of VAMP2/VAMP3 vesicles in astrocyte signaling. We also discuss our recent data suggesting that astrocytic VAMP3 vesicles regulate the trafficking of glutamate transporters at the plasma membrane and glutamate uptake. A better understanding of the functional consequences of the astrocytic vSNARE vesicles on glutamate signaling, neuronal excitability and plasticity, will require the development of new strategies to selectively interrogate the astrocytic vesicles trafficking in vivo.
Collapse
Affiliation(s)
- N Ropert
- Brain Physiology Laboratory, CNRS UMR8118, Paris F-75006, France; Fédération de Recherche en Neurosciences, FR 3636, Université Paris Descartes, 45 rue des Saints Pères, Paris F-75006, France; Sorbonne Paris Cité, 190, avenue de France, Paris F-75013, France
| | - A Jalil
- Brain Physiology Laboratory, CNRS UMR8118, Paris F-75006, France; Fédération de Recherche en Neurosciences, FR 3636, Université Paris Descartes, 45 rue des Saints Pères, Paris F-75006, France; Sorbonne Paris Cité, 190, avenue de France, Paris F-75013, France
| | - D Li
- Brain Physiology Laboratory, CNRS UMR8118, Paris F-75006, France; Fédération de Recherche en Neurosciences, FR 3636, Université Paris Descartes, 45 rue des Saints Pères, Paris F-75006, France; Sorbonne Paris Cité, 190, avenue de France, Paris F-75013, France.
| |
Collapse
|
34
|
Sadler JBA, Roccisana J, Virolainen M, Bryant NJ, Gould GW. mVps45 knockdown selectively modulates VAMP expression in 3T3-L1 adipocytes. Commun Integr Biol 2015; 8:e1026494. [PMID: 26479872 PMCID: PMC4594494 DOI: 10.1080/19420889.2015.1026494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 08/05/2014] [Accepted: 08/06/2014] [Indexed: 02/08/2023] Open
Abstract
Insulin stimulates the delivery of glucose transporter-4 (GLUT4)-containing vesicles to the surface of adipocytes. Depletion of the Sec1/Munc18 protein mVps45 significantly abrogates insulin-stimulated glucose transport and GLUT4 translocation. Here we show that depletion of mVps45 selectively reduced expression of VAMPs 2 and 4, but not other VAMP isoforms. Although we did not observe direct interaction of mVps45 with any VAMP isoform; we found that the cognate binding partner of mVps45, Syntaxin 16 associates with VAMPs 2, 4, 7 and 8 in vitro. Co-immunoprecipitation experiments in 3T3-L1 adipocytes revealed an interaction between Syntaxin 16 and only VAMP4. We suggest GLUT4 trafficking is controlled by the coordinated expression of mVps45/Syntaxin 16/VAMP4, and that depletion of mVps45 regulates VAMP2 levels indirectly, perhaps via reduced trafficking into specialized subcellular compartments.
Collapse
Affiliation(s)
- Jessica B A Sadler
- Henry Wellcome Laboratory of Cell Biology; Institute of Molecular; Cell and Systems Biology; College of Medical; Veterinary and Life Sciences; University of Glasgow ; Glasgow, Scotland
| | - Jennifer Roccisana
- Henry Wellcome Laboratory of Cell Biology; Institute of Molecular; Cell and Systems Biology; College of Medical; Veterinary and Life Sciences; University of Glasgow ; Glasgow, Scotland
| | - Minttu Virolainen
- Henry Wellcome Laboratory of Cell Biology; Institute of Molecular; Cell and Systems Biology; College of Medical; Veterinary and Life Sciences; University of Glasgow ; Glasgow, Scotland
| | - Nia J Bryant
- Department of Biology; University of York ; Heslington, York
| | - Gwyn W Gould
- Henry Wellcome Laboratory of Cell Biology; Institute of Molecular; Cell and Systems Biology; College of Medical; Veterinary and Life Sciences; University of Glasgow ; Glasgow, Scotland
| |
Collapse
|
35
|
Renard HF, Garcia-Castillo MD, Chambon V, Lamaze C, Johannes L. Shiga toxin stimulates clathrin-independent endocytosis of the VAMP2, VAMP3 and VAMP8 SNARE proteins. J Cell Sci 2015; 128:2891-902. [PMID: 26071526 DOI: 10.1242/jcs.171116] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 06/08/2015] [Indexed: 01/08/2023] Open
Abstract
Endocytosis is an essential cellular process that is often hijacked by pathogens and pathogenic products. Endocytic processes can be classified into two broad categories, those that are dependent on clathrin and those that are not. The SNARE proteins VAMP2, VAMP3 and VAMP8 are internalized in a clathrin-dependent manner. However, the full scope of their endocytic behavior has not yet been elucidated. Here, we found that VAMP2, VAMP3 and VAMP8 are localized on plasma membrane invaginations and very early uptake structures that are induced by the bacterial Shiga toxin, which enters cells by clathrin-independent endocytosis. We show that toxin trafficking into cells and cell intoxication rely on these SNARE proteins. Of note, the cellular uptake of VAMP3 is increased in the presence of Shiga toxin, even when clathrin-dependent endocytosis is blocked. We therefore conclude that VAMP2, VAMP3 and VAMP8 are removed from the plasma membrane by non-clathrin-mediated pathways, in addition to by clathrin-dependent uptake. Moreover, our study identifies these SNARE proteins as the first transmembrane trafficking factors that functionally associate at the plasma membrane with the toxin-driven clathrin-independent invaginations during the uptake process.
Collapse
Affiliation(s)
- Henri-François Renard
- Institut Curie - Centre de Recherche, Endocytic Trafficking and Therapeutic Delivery Group, 26 rue d'Ulm, Paris 75248, Cedex 05, France CNRS UMR3666, Paris 75005, France INSERM U1143, Paris 75005, France
| | - Maria Daniela Garcia-Castillo
- Institut Curie - Centre de Recherche, Endocytic Trafficking and Therapeutic Delivery Group, 26 rue d'Ulm, Paris 75248, Cedex 05, France CNRS UMR3666, Paris 75005, France INSERM U1143, Paris 75005, France
| | - Valérie Chambon
- Institut Curie - Centre de Recherche, Endocytic Trafficking and Therapeutic Delivery Group, 26 rue d'Ulm, Paris 75248, Cedex 05, France CNRS UMR3666, Paris 75005, France INSERM U1143, Paris 75005, France
| | - Christophe Lamaze
- CNRS UMR3666, Paris 75005, France INSERM U1143, Paris 75005, France Institut Curie - Centre de Recherche, Membrane Dynamics and Mechanics of Intracellular Signaling Group, 26 rue d'Ulm, Paris 75248, Cedex 05, France
| | - Ludger Johannes
- Institut Curie - Centre de Recherche, Endocytic Trafficking and Therapeutic Delivery Group, 26 rue d'Ulm, Paris 75248, Cedex 05, France CNRS UMR3666, Paris 75005, France INSERM U1143, Paris 75005, France
| |
Collapse
|
36
|
Sadler JBA, Bryant NJ, Gould GW. Characterization of VAMP isoforms in 3T3-L1 adipocytes: implications for GLUT4 trafficking. Mol Biol Cell 2014; 26:530-6. [PMID: 25501368 PMCID: PMC4310743 DOI: 10.1091/mbc.e14-09-1368] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The levels of expression, distribution, and association of all of the VAMPs expressed in 3T3-L1 adipocytes are characterized. This is the first systematic analysis of all members of this protein family for any cell type. The fusion of GLUT4-containing vesicles with the plasma membrane of adipocytes is a key facet of insulin action. This process is mediated by the formation of functional soluble N-ethylmaleimide–sensitive factor attachment protein receptor (SNARE) complexes between the plasma membrane t-SNARE complex and the vesicle v-SNARE or VAMP. The t-SNARE complex consists of Syntaxin4 and SNAP23, and whereas many studies identify VAMP2 as the v-SNARE, others suggest that either VAMP3 or VAMP8 may also fulfil this role. Here we characterized the levels of expression, distribution, and association of all the VAMPs expressed in 3T3-L1 adipocytes to provide the first systematic analysis of all members of this protein family for any cell type. Despite our finding that all VAMP isoforms form SDS-resistant SNARE complexes with Syntaxin4/SNAP23 in vitro, a combination of levels of expression (which vary by >30-fold), subcellular distribution, and coimmunoprecipitation analyses lead us to propose that VAMP2 is the major v-SNARE involved in GLUT4 trafficking to the surface of 3T3-L1 adipocytes.
Collapse
Affiliation(s)
- Jessica B A Sadler
- Henry Wellcome Laboratory of Cell Biology, Institute for Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Nia J Bryant
- Department of Biology, University of York, Heslington YO10 5DD, United Kingdom
| | - Gwyn W Gould
- Henry Wellcome Laboratory of Cell Biology, Institute for Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| |
Collapse
|
37
|
Organization of organelles and VAMP-associated vesicular transport systems in differentiating skeletal muscle cells. Anat Sci Int 2014; 90:33-9. [DOI: 10.1007/s12565-014-0266-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 11/19/2014] [Indexed: 10/24/2022]
|
38
|
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]
|
39
|
Li J, Cantley J, Burchfield JG, Meoli CC, Stöckli J, Whitworth PT, Pant H, Chaudhuri R, Groffen AJA, Verhage M, James DE. DOC2 isoforms play dual roles in insulin secretion and insulin-stimulated glucose uptake. Diabetologia 2014; 57:2173-82. [PMID: 25005332 DOI: 10.1007/s00125-014-3312-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 05/28/2014] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS Glucose-stimulated insulin secretion (GSIS) and insulin-stimulated glucose uptake are processes that rely on regulated intracellular vesicle transport and vesicle fusion with the plasma membrane. DOC2A and DOC2B are calcium-sensitive proteins that were identified as key components of vesicle exocytosis in neurons. Our aim was to investigate the role of DOC2 isoforms in glucose homeostasis, insulin secretion and insulin action. METHODS DOC2 expression was measured by RT-PCR and western blotting. Body weight, glucose tolerance, insulin action and GSIS were assessed in wild-type (WT), Doc2a (-/-) (Doc2aKO), Doc2b (-/-) (Doc2bKO) and Doc2a (-/-)/Doc2b (-/-) (Doc2a/Doc2bKO) mice in vivo. In vitro GSIS and glucose uptake were assessed in isolated tissues, and exocytotic proteins measured by western blotting. GLUT4 translocation was assessed by epifluorescence microscopy. RESULTS Doc2b mRNA was detected in all tissues tested, whereas Doc2a was only detected in islets and the brain. Doc2aKO and Doc2bKO mice had minor glucose intolerance, while Doc2a/Doc2bKO mice showed pronounced glucose intolerance. GSIS was markedly impaired in Doc2a/Doc2bKO mice in vivo, and in isolated Doc2a/Doc2bKO islets in vitro. In contrast, Doc2bKO mice had only subtle defects in insulin secretion in vivo. Insulin action was impaired to a similar degree in both Doc2bKO and Doc2a/Doc2bKO mice. In vitro insulin-stimulated glucose transport and GLUT4 vesicle fusion were defective in adipocytes derived from Doc2bKO mice. Surprisingly, insulin action was not altered in muscle isolated from DOC2-null mice. CONCLUSIONS/INTERPRETATION Our study identifies a critical role for DOC2B in insulin-stimulated glucose uptake in adipocytes, and for the synergistic regulation of GSIS by DOC2A and DOC2B in beta cells.
Collapse
Affiliation(s)
- Jia Li
- Diabetes and Obesity Research Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
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
|
41
|
Chen Y, Meng D, Wang H, Sun R, Wang D, Wang S, Fan J, Zhao Y, Wang J, Yang S, Huai C, Song X, Qin R, Xu T, Yun D, Hu L, Yang J, Zhang X, Chen H, Chen J, Chen H, Lu D. VAMP8 facilitates cellular proliferation and temozolomide resistance in human glioma cells. Neuro Oncol 2014; 17:407-18. [PMID: 25209430 DOI: 10.1093/neuonc/nou219] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 07/20/2014] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Malignant glioma is a common and lethal primary brain tumor in adults. Here we identified a novel oncoprotein, vesicle-associated membrane protein 8 (VAMP8), and investigated its roles in tumorigenisis and chemoresistance in glioma. METHODS The expression of gene and protein were determined by quantitative PCR and Western blot, respectively. Histological analysis of 282 glioma samples and 12 normal controls was performed by Pearson's chi-squared test. Survival analysis was performed using the log-rank test and Cox proportional hazards regression. Cell proliferation and cytotoxicity assay were conducted using Cell Counting Kit-8. Autophagy was detected by confocal microscopy and Western blot. RESULTS VAMP8 was significantly overexpressed in human glioma specimens and could become a potential novel prognostic and treatment-predictive marker for glioma patients. Overexpression of VAMP8 promoted cell proliferation in vitro and in vivo, whereas knockdown of VAMP8 attenuated glioma growth by arresting cell cycle in the G0/G1 phase. Moreover, VAMP8 contributed to temozolomide (TMZ) resistance by elevating the expression levels of autophagy proteins and the number of autophagosomes. Further inhibition of autophagy via siRNA-mediated knockdown of autophagy-related gene 5 (ATG5) or syntaxin 17 (STX17) reversed TMZ resistance in VAMP8-overexpressing cells, while silencing of VAMP8 impaired the autophagic flux and alleviated TMZ resistance in glioma cells. CONCLUSION Our findings identified VAMP8 as a novel oncogene by promoting cell proliferation and therapeutic resistance in glioma. Targeting VAMP8 may serve as a potential therapeutic regimen for the treatment of glioma.
Collapse
Affiliation(s)
- Yuanyuan Chen
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Delong Meng
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Huibo Wang
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Ruochuan Sun
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Dongrui Wang
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Shuai Wang
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Jiajun Fan
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Yingjie Zhao
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Jingkun Wang
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Song Yang
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Cong Huai
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Xiao Song
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Rong Qin
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Tao Xu
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Dapeng Yun
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Lingna Hu
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Jingmin Yang
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Xiaotian Zhang
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Haoming Chen
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Juxiang Chen
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Hongyan Chen
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| | - Daru Lu
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Shanghai, China (Y.C., D.M., D.W., Y.Z., J.W., C.H., X.S., D.Y., L.H., J.Y., H.C., H.C., D.L.); Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China (J.F.); Department of Neurosurgery, (H.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China (S.W.); Eighth Department of General Surgery and Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, China (R.S., S.Y.); Department of Molecular Human Genetics, Baylor College of Medicine, Houston, Texas (X.Z.); Neurosurgery Research Institution of Shanghai, Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China (R.Q., T.X., J.C.)
| |
Collapse
|
42
|
Gaisano HY. Here come the newcomer granules, better late than never. Trends Endocrinol Metab 2014; 25:381-8. [PMID: 24746186 DOI: 10.1016/j.tem.2014.03.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 03/06/2014] [Accepted: 03/14/2014] [Indexed: 01/03/2023]
Abstract
Exocytosis in pancreatic β-cells employs Munc18/soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes that mediate the priming and docking onto the plasma membrane (PM) of insulin granules, called predocked granules, that sit on the PM until Ca(2+) influx evokes fusion. This accounts for most of the initial peak secretory response. However, the subsequent sustained phase of glucose-stimulated insulin secretion arises from newcomer granules that have a minimal residence time at the PM before fusion. In this Opinion I discuss recent work that has begun to decipher the components of the exocytotic machinery of newcomer granules, including a Munc18/SNARE complex that is different from that mediating the fusion of predocked granules and which can potentially rescue defective insulin secretion in diabetes. These insights are applicable to other neuroendocrine cells that exhibit sustained secretion.
Collapse
Affiliation(s)
- Herbert Y Gaisano
- Department of Medicine, University of Toronto, M5S 1A8, Toronto, Canada.
| |
Collapse
|
43
|
Alfadda AA, Turjoman AA, Moustafa AS, Al-Naami MY, Chishti MA, Sallam RM, Gibson D, Duncan MW. A proteomic analysis of excreted and circulating proteins from obese patients following two different weight-loss strategies. Exp Biol Med (Maywood) 2014; 239:568-80. [PMID: 24668551 DOI: 10.1177/1535370214523894] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Bariatric surgery is the most successful therapeutic approach to weight loss, but how it leads to weight loss, and how it resolves obesity-related complications, including type-2 diabetes, are poorly understood. This study, comprising two groups of individuals, one on a low-calorie diet (n = 5) and one undergoing bariatric surgery (n = 7), used both targeted and untargeted proteomic approaches to determine changes in protein levels pre- and post-intervention (i.e. 3-6 months later). Changes were observed in both circulating and excreted proteins following weight loss. Targeted multiplexed biochip arrays measured 12 plasma peptides/proteins involved in metabolism and inflammation: C-peptide, ferritin, interleukin-6, interleukin-1 alpha, resistin, insulin, tumor necrosis factor alpha, leptin, plasminogen-activator inhibitor-1, adiponectin, cystatin C, and C-reactive protein. Following a low-calorie diet, plasma insulin and C-reactive protein levels were significantly reduced (P = 0.045 and P = 0.030, respectively); adiponectin increased and leptin decreased following surgery (P = 0.014 and P = 0.005, respectively). Untargeted proteomic analysis employing 2D difference in-gel electrophoresis (DIGE) showed 28 protein spots with ≥1.5-fold changes in expression following weight loss by a low-calorie diet; comparison of pre- and post-intervention urine samples from the bariatric surgery group showed changes in excretion of 110 protein spots. The combination of targeted protein analysis by multiplexed arrays and an exploratory (i.e. an unbiased or discovery) proteomic assessment of hundreds of proteins offers valuable insights into the mechanistic differences between alternative weight-loss strategies. This is a powerful hypothesis-generating approach to study complex, multifactorial syndromes such as obesity. The findings that arise from these studies can then be validated in targeted, hypothesis-directed investigations.
Collapse
Affiliation(s)
- Assim A Alfadda
- Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | | | | | | | | | | | | | | |
Collapse
|
44
|
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
|
45
|
Yang P, Patrick E, Tan SX, Fazakerley DJ, Burchfield J, Gribben C, Prior MJ, James DE, Hwa Yang Y. Direction pathway analysis of large-scale proteomics data reveals novel features of the insulin action pathway. ACTA ACUST UNITED AC 2013; 30:808-14. [PMID: 24167158 DOI: 10.1093/bioinformatics/btt616] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
MOTIVATION With the advancement of high-throughput techniques, large-scale profiling of biological systems with multiple experimental perturbations is becoming more prevalent. Pathway analysis incorporates prior biological knowledge to analyze genes/proteins in groups in a biological context. However, the hypotheses under investigation are often confined to a 1D space (i.e. up, down, either or mixed regulation). Here, we develop direction pathway analysis (DPA), which can be applied to test hypothesis in a high-dimensional space for identifying pathways that display distinct responses across multiple perturbations. RESULTS Our DPA approach allows for the identification of pathways that display distinct responses across multiple perturbations. To demonstrate the utility and effectiveness, we evaluated DPA under various simulated scenarios and applied it to study insulin action in adipocytes. A major action of insulin in adipocytes is to regulate the movement of proteins from the interior to the cell surface membrane. Quantitative mass spectrometry-based proteomics was used to study this process on a large-scale. The combined dataset comprises four separate treatments. By applying DPA, we identified that several insulin responsive pathways in the plasma membrane trafficking are only partially dependent on the insulin-regulated kinase Akt. We subsequently validated our findings through targeted analysis of key proteins from these pathways using immunoblotting and live cell microscopy. Our results demonstrate that DPA can be applied to dissect pathway networks testing diverse hypotheses and integrating multiple experimental perturbations. AVAILABILITY AND IMPLEMENTATION The R package 'directPA' is distributed from CRAN under GNU General Public License (GPL)-3 and can be downloaded from: http://cran.r-project.org/web/packages/directPA/index.html CONTACT jean.yang@sydney.edu.au SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Pengyi Yang
- Systems Biology Group, Biostatistics Branch, National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, NC 27709, USA, School of Mathematics and Statistics, University of Sydney, Diabetes and Obesity Program, Garvan Institute of Medical Research, NSW 2006, Australia and Metabolism in Human Disease Unit, Institute of Molecular and Cellular Biology, A*Star, 61 Biopolis Drive, Proteos 138673, Singapore
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Lopez JA, Voskoboinik I. Deciphering the syntax of cytotoxic lymphocyte degranulation. Eur J Immunol 2013; 43:46-9. [PMID: 23322694 DOI: 10.1002/eji.201243221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 12/03/2012] [Accepted: 12/10/2012] [Indexed: 11/09/2022]
Abstract
In the killer lymphocyte, the targeted delivery of perforin- and granzyme-containing cytotoxic granules to the immunological synapse is crucial for the eradication of pathogen-infected or transformed targets. This process is achieved through a tightly controlled and highly efficient granule exocytosis pathway. Mutations in the granule trafficking proteins Munc13-4, syntaxin 11, Munc18-2 or Rab27 leads to a fatal lapse of immune surveillance and can be manifested as haemophagocytic lymphohistiocytosis in humans. Elucidation of the role of these proteins in exocytic trafficking is pivotal for our understanding of their role in health and disease. In this issue of the European Journal of Immunology, D'Orlando et al. [Eur. J. Immunol. 2013. 43: 194-208] make an important step in this direction, as they generate and characterise syntaxin 11 deficient mice. Herein, we discuss the role of syntaxin-11 in soluble NSF (N-ethylmaleimide sensitive fusion) attachment protein receptors complex formation leading to cytotoxic lymphocyte degranulation and its importance in maintaining immune homeostasis.
Collapse
Affiliation(s)
- Jamie A Lopez
- Cancer Immunology Program, Peter MacCallum Cancer Centre, St. Andrews Place, East Melbourne, Australia
| | | |
Collapse
|
47
|
Yu H, Rathore SS, Davis EM, Ouyang Y, Shen J. Doc2b promotes GLUT4 exocytosis by activating the SNARE-mediated fusion reaction in a calcium- and membrane bending-dependent manner. Mol Biol Cell 2013; 24:1176-84. [PMID: 23427263 PMCID: PMC3623638 DOI: 10.1091/mbc.e12-11-0810] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Reconstitution of GLUT4 vesicle fusion in a defined fusion system shows that the C2-domain factor Doc2b activates the SNARE-dependent fusion reaction by a calcium- and membrane bending–dependent mechanism. Of importance, certain features of Doc2b function appear to be distinct from how synaptotagmin-1 promotes synaptic release. The glucose transporter GLUT4 plays a central role in maintaining body glucose homeostasis. On insulin stimulation, GLUT4-containing vesicles fuse with the plasma membrane, relocating GLUT4 from intracellular reservoirs to the cell surface to uptake excess blood glucose. The GLUT4 vesicle fusion reaction requires soluble N-ethylmaleimide–sensitive factor attachment protein receptors (SNAREs) as the core fusion engine and a group of regulatory proteins. In particular, the soluble C2-domain factor Doc2b plays a key role in GLUT4 vesicle fusion, but its molecular mechanism has been unclear. Here we reconstituted the SNARE-dependent GLUT4 vesicle fusion in a defined proteoliposome fusion system. We observed that Doc2b binds to GLUT4 exocytic SNAREs and potently accelerates the fusion kinetics in the presence of Ca2+. The stimulatory activity of Doc2b requires intact Ca2+-binding sites on both the C2A and C2B domains. Using electron microscopy, we observed that Doc2b strongly bends the membrane bilayer, and this membrane-bending activity is essential to the stimulatory function of Doc2b in fusion. These results demonstrate that Doc2b promotes GLUT4 exocytosis by accelerating the SNARE-dependent fusion reaction by a Ca2+- and membrane bending–dependent mechanism. Of importance, certain features of Doc2b function appear to be distinct from how synaptotagmin-1 promotes synaptic neurotransmitter release, suggesting that exocytic Ca2+ sensors may possess divergent mechanisms in regulating vesicle fusion.
Collapse
Affiliation(s)
- Haijia Yu
- Department of Molecular, Cellular and Developmental Biology, University of Colorado at Boulder, Boulder, CO 80309, USA
| | | | | | | | | |
Collapse
|
48
|
Burchfield JG, Lu J, Fazakerley DJ, Tan SX, Ng Y, Mele K, Buckley MJ, Han W, Hughes WE, James DE. Novel systems for dynamically assessing insulin action in live cells reveals heterogeneity in the insulin response. Traffic 2013; 14:259-73. [PMID: 23252720 DOI: 10.1111/tra.12035] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 12/13/2012] [Accepted: 12/18/2012] [Indexed: 12/23/2022]
Abstract
Regulated GLUT4 trafficking is a key action of insulin. Quantitative stepwise analysis of this process provides a powerful tool for pinpointing regulatory nodes that contribute to insulin regulation and insulin resistance. We describe a novel GLUT4 construct and workflow for the streamlined dissection of GLUT4 trafficking; from simple high throughput screens to high resolution analyses of individual vesicles. We reveal single cell heterogeneity in insulin action highlighting the utility of this approach - each cell displayed a unique and highly reproducible insulin response, implying that each cell is hard-wired to produce a specific output in response to a given stimulus. These data highlight that the response of a cell population to insulin is underpinned by extensive heterogeneity at the single cell level. This heterogeneity is pre-programmed within each cell and is not the result of intracellular stochastic events.
Collapse
Affiliation(s)
- James G Burchfield
- Diabetes and Obesity Research Program, The Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
|
50
|
Zhu D, Zhang Y, Lam PPL, Dolai S, Liu Y, Cai EP, Choi D, Schroer SA, Kang Y, Allister EM, Qin T, Wheeler MB, Wang CC, Hong WJ, Woo M, Gaisano HY. Dual role of VAMP8 in regulating insulin exocytosis and islet β cell growth. Cell Metab 2012; 16:238-49. [PMID: 22841572 DOI: 10.1016/j.cmet.2012.07.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 04/17/2012] [Accepted: 06/26/2012] [Indexed: 12/22/2022]
Abstract
Optimal insulin secretion required to maintain glucose homeostasis is the summation of total pancreatic islet β cell mass and intrinsic secretory capacity of individual β cells, which are regulated by distinct mechanisms that could be amplified by glucagon-like-peptide-1 (GLP-1). Because of these actions of GLP-1 on islet β cells, GLP-1 has been deployed to treat diabetes. We employed SNARE protein VAMP8-null mice to demonstrate that VAMP8 mediates insulin granule recruitment to the plasma membrane, which partly accounts for GLP-1 potentiation of glucose-stimulated insulin secretion. VAMP8-null mice also exhibited increased islet β cell mass from increased β cell mitosis, with β cell proliferative activity greatly amplified by GLP-1. Thus, despite the β cell exocytotic defect, VAMP8-null mice have an increased total insulin secretory capacity, which improved glucose homeostasis. We conclude that these VAMP8-mediated events partly underlie the therapeutic actions of GLP-1 on insulin secretion and β cell growth.
Collapse
Affiliation(s)
- Dan Zhu
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|