1
|
Chen YC, Bäck NE, Zhen J, Xiong L, Komba M, Gloyn AL, MacDonald PE, Mains RE, Eipper BA, Verchere CB. Peptidylglycine alpha-amidating monooxygenase is important in mice for beta-cell cilia formation and insulin secretion but promotes diabetes risk through beta-cell independent mechanisms. Mol Metab 2025; 96:102123. [PMID: 40120979 DOI: 10.1016/j.molmet.2025.102123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/10/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
OBJECTIVES Carriers of PAM (peptidylglycine alpha-amidating monooxygenase) coding variant alleles have reduced insulinogenic index, higher risk of developing type 2 diabetes (T2D), and islets from heterozygous carriers of the PAM p.Asp563Gly variant display reduced insulin secretion. Exactly how global PAM deficiency contributes to hyperglycemia remains unclear. PAM is the only enzyme capable of converting glycine-extended peptide hormones into amidated products. Like neuropeptide Y (NPY), α-melanocyte stimulating hormone (αMSH), and glucagon-like peptide 1 (GLP-1), islet amyloid polypeptide (IAPP), a beta cell peptide that forms islet amyloid in type 2 diabetes, is a PAM substrate. We hypothesized that Pam deficiency limited to beta cells would lead to reduced insulin secretion, prevent the production of amidated IAPP, and reveal the extent to which loss of Pam in β-cells could accelerate the onset of hyperglycemia in mice. METHODS PAM activity was assessed in human islets from donors based on their PAM genotype. We generated beta cell-specific Pam knockout (Ins1Cre/+, Pamfl/fl; βPamKO) mice and performed islet culture, histological, and metabolic assays to evaluate the physiological roles of Pam in beta cells. We analyzed human IAPP (hIAPP) amyloid fibril forming kinetics using synthetic amidated and non-amidated hIAPP peptides, and generated hIAPP knock-in beta cell-specific Pam knockout (hIAPPw/w βPamKO) mice to determine the impact of hIAPP amidation on islet amyloid burden, islet graft survival, and glucose tolerance. RESULTS PAM enzyme activity was significantly reduced in islets from donors with the PAM p. Asp563Gly T2D-risk allele. Islets from βPamKO mice had impaired second-phase glucose- and KCl-induced insulin secretion. Beta cells from βPamKO mice had larger dense-core granules and fewer and shorter cilia. Interestingly, non-amidated hIAPP was less fibrillogenic in vitro, and high glucose-treated hIAPPw/w βPamKO islets had reduced amyloid burden. Despite these changes in beta cell function, βPamKO mice were not more susceptible to diet-induced hyperglycemia. In vitro beta cell death and in vivo islet graft survival remained comparable between hIAPPw/w βPamKO and hIAPPw/w islets. Surprisingly, aged hIAPPw/w βPamKO mice had improved insulin secretion and glucose tolerance. CONCLUSIONS Eliminating Pam expression only in beta cells leads to morphological changes in insulin granules, reduced insulin secretion, reduced hIAPP amyloid burden and altered ciliogenesis. However, in mice beta-cell Pam deficiency has no impact on the development of diet- or hIAPP-induced hyperglycemia. Our data are consistent with current studies revealing ancient, highly conserved roles for peptidergic signaling in the coordination of the diverse signals needed to regulate fundamental processes such as glucose homeostasis.
Collapse
Affiliation(s)
- Yi-Chun Chen
- Department of Surgery, Faculty of Medicine, University of British Columbia & BC Children's Hospital Research Institute, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada.
| | - Nils E Bäck
- Department of Anatomy, Faculty of Medicine, University of Helsinki, PO Box 63 (Haartmaninkatu 8), 00014 University of Helsinki, Finland.
| | - Jenicia Zhen
- Department of Surgery, Faculty of Medicine, University of British Columbia & BC Children's Hospital Research Institute, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada.
| | - Lena Xiong
- Department of Surgery, Faculty of Medicine, University of British Columbia & BC Children's Hospital Research Institute, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
| | - Mitsuhiro Komba
- Department of Surgery, Faculty of Medicine, University of British Columbia & BC Children's Hospital Research Institute, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada.
| | - Anna L Gloyn
- Department of Pediatrics, Division of Endocrinology & Diabetes and Department of Genetics, Stanford School of Medicine, Stanford Research Park, 3165 Porter Drive, Stanford, CA, 94304, USA.
| | - Patrick E MacDonald
- Department of Pharmacology and Alberta Diabetes Institute, 6-126C Li Ka Shing Centre for Health Research Innovation, Alberta Diabetes Institute, University of Alberta, Edmonton, AB, T6G 2E1, Canada.
| | - Richard E Mains
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, MC 3401, Farmington, CT, 06030-3401, USA.
| | - Betty A Eipper
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, MC 3401, Farmington, CT, 06030-3401, USA.
| | - C Bruce Verchere
- Department of Surgery, Faculty of Medicine, University of British Columbia & BC Children's Hospital Research Institute, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada; Centre for Molecular Medicine and Therapeutics, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada.
| |
Collapse
|
2
|
Xie L, Kang F, Qin T, Kang Y, Liang T, Xie H, Froese CD, Xie H, Au A, Yip CM, Trimble WS, Gaisano HY. Septin5 deletion enhances β-cell exocytosis by releasing microtubule-tethered insulin granules onto plasma membrane. Nat Commun 2025; 16:2725. [PMID: 40108136 PMCID: PMC11923188 DOI: 10.1038/s41467-025-57421-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
Septin5 interacts with SNARE proteins to regulate exocytosis in neurons, but its role in pancreatic β-cells is unknown. Here, we report that Septin5 is abundant in rodent and human β-cells, deletion of which dramatically enhances biphasic glucose-stimulated insulin secretion, including in type 2 diabetes (T2D). Super-resolution imaging shows that Septin5 is preferentially assembled in microtubule-plasma membrane contact sites in a microtubule-dependent manner, which provides discrete harbor for secretory granule anchoring. By decreasing the stability of the cortical microtubule meshwork, Septin5 depletion increases insulin granule dynamics and access to the plasma membrane. Analysis of spatiotemporal coupling of fusion events and localized Ca2+ influx through L-type Ca2+ channels show that Septin5 depletion increases releasable granule pool clustering on Ca2+ channels, previously shown to be impaired in T2D, thus rectifying this T2D defect. Hence, inhibition of Septin5 can improve insulin secretion.
Collapse
Affiliation(s)
- Li Xie
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.
| | - Fei Kang
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.
| | - Tairan Qin
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Youhou Kang
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Tao Liang
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Huanli Xie
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Carol D Froese
- The program in Cell Biology, Department of Biochemistry, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Hong Xie
- The program in Cell Biology, Department of Biochemistry, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Aaron Au
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Christopher M Yip
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - William S Trimble
- The program in Cell Biology, Department of Biochemistry, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Herbert Y Gaisano
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
3
|
Szodorai E, Hevesi Z, Wagner L, Hökfelt TGM, Harkany T, Schnell R. A hydrophobic groove in secretagogin allows for alternate interactions with SNAP-25 and syntaxin-4 in endocrine tissues. Proc Natl Acad Sci U S A 2024; 121:e2309211121. [PMID: 38593081 PMCID: PMC11032447 DOI: 10.1073/pnas.2309211121] [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: 06/01/2023] [Accepted: 03/09/2024] [Indexed: 04/11/2024] Open
Abstract
Vesicular release of neurotransmitters and hormones relies on the dynamic assembly of the exocytosis/trans-SNARE complex through sequential interactions of synaptobrevins, syntaxins, and SNAP-25. Despite SNARE-mediated release being fundamental for intercellular communication in all excitable tissues, the role of auxiliary proteins modulating the import of reserve vesicles to the active zone, and thus, scaling repetitive exocytosis remains less explored. Secretagogin is a Ca2+-sensor protein with SNAP-25 being its only known interacting partner. SNAP-25 anchors readily releasable vesicles within the active zone, thus being instrumental for 1st phase release. However, genetic deletion of secretagogin impedes 2nd phase release instead, calling for the existence of alternative protein-protein interactions. Here, we screened the secretagogin interactome in the brain and pancreas, and found syntaxin-4 grossly overrepresented. Ca2+-loaded secretagogin interacted with syntaxin-4 at nanomolar affinity and 1:1 stoichiometry. Crystal structures of the protein complexes revealed a hydrophobic groove in secretagogin for the binding of syntaxin-4. This groove was also used to bind SNAP-25. In mixtures of equimolar recombinant proteins, SNAP-25 was sequestered by secretagogin in competition with syntaxin-4. Kd differences suggested that secretagogin could shape unidirectional vesicle movement by sequential interactions, a hypothesis supported by in vitro biological data. This mechanism could facilitate the movement of transport vesicles toward release sites, particularly in the endocrine pancreas where secretagogin, SNAP-25, and syntaxin-4 coexist in both α- and β-cells. Thus, secretagogin could modulate the pace and fidelity of vesicular hormone release by differential protein interactions.
Collapse
Affiliation(s)
- Edit Szodorai
- Division of Molecular and Cellular Neuroendocrinology, Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, SolnaSE-17165, Sweden
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, ViennaA-1090, Austria
| | - Zsofia Hevesi
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, ViennaA-1090, Austria
| | - Ludwig Wagner
- Department of Internal Medicine III, Medical University of Vienna, ViennaA-1090, Austria
| | - Tomas G. M. Hökfelt
- Division of Molecular and Cellular Neuroendocrinology, Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, SolnaSE-17165, Sweden
| | - Tibor Harkany
- Division of Molecular and Cellular Neuroendocrinology, Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, SolnaSE-17165, Sweden
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, ViennaA-1090, Austria
| | - Robert Schnell
- Division of Molecular and Cellular Neuroendocrinology, Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, SolnaSE-17165, Sweden
| |
Collapse
|
4
|
Rahman MM, Pathak A, Schueler KL, Alsharif H, Michl A, Alexander J, Kim JA, Bhatnagar S. Genetic ablation of synaptotagmin-9 alters tomosyn-1 function to increase insulin secretion from pancreatic β-cells improving glucose clearance. FASEB J 2023; 37:e23075. [PMID: 37432648 PMCID: PMC10348599 DOI: 10.1096/fj.202300291rr] [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: 02/18/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/12/2023]
Abstract
Stimulus-coupled insulin secretion from the pancreatic islet β-cells involves the fusion of insulin granules to the plasma membrane (PM) via SNARE complex formation-a cellular process key for maintaining whole-body glucose homeostasis. Less is known about the role of endogenous inhibitors of SNARE complexes in insulin secretion. We show that an insulin granule protein synaptotagmin-9 (Syt9) deletion in mice increased glucose clearance and plasma insulin levels without affecting insulin action compared to the control mice. Upon glucose stimulation, increased biphasic and static insulin secretion were observed from ex vivo islets due to Syt9 loss. Syt9 colocalizes and binds with tomosyn-1 and the PM syntaxin-1A (Stx1A); Stx1A is required for forming SNARE complexes. Syt9 knockdown reduced tomosyn-1 protein abundance via proteasomal degradation and binding of tomosyn-1 to Stx1A. Furthermore, Stx1A-SNARE complex formation was increased, implicating Syt9-tomosyn-1-Stx1A complex is inhibitory in insulin secretion. Rescuing tomosyn-1 blocked the Syt9-knockdown-mediated increases in insulin secretion. This shows that the inhibitory effects of Syt9 on insulin secretion are mediated by tomosyn-1. We report a molecular mechanism by which β-cells modulate their secretory capacity rendering insulin granules nonfusogenic by forming the Syt9-tomosyn-1-Stx1A complex. Altogether, Syt9 loss in β-cells decreases tomosyn-1 protein abundance, increasing the formation of Stx1A-SNARE complexes, insulin secretion, and glucose clearance. These outcomes differ from the previously published work that identified Syt9 has either a positive or no effect of Syt9 on insulin secretion. Future work using β-cell-specific deletion of Syt9 mice is key for establishing the role of Syt9 in insulin secretion.
Collapse
Affiliation(s)
- Md Mostafizur Rahman
- Heersink School of Medicine, Division of Endocrinology, Diabetes, & Metabolism, Comprehensive Diabetes Center, University of Alabama, Birmingham, AL, 35294
| | - Asmita Pathak
- Heersink School of Medicine, Division of Endocrinology, Diabetes, & Metabolism, Comprehensive Diabetes Center, University of Alabama, Birmingham, AL, 35294
| | | | - Haifa Alsharif
- Heersink School of Medicine, Division of Endocrinology, Diabetes, & Metabolism, Comprehensive Diabetes Center, University of Alabama, Birmingham, AL, 35294
| | - Ava Michl
- Heersink School of Medicine, Division of Endocrinology, Diabetes, & Metabolism, Comprehensive Diabetes Center, University of Alabama, Birmingham, AL, 35294
| | - Justin Alexander
- Heersink School of Medicine, Division of Endocrinology, Diabetes, & Metabolism, Comprehensive Diabetes Center, University of Alabama, Birmingham, AL, 35294
| | - Jeong-A Kim
- Heersink School of Medicine, Division of Endocrinology, Diabetes, & Metabolism, Comprehensive Diabetes Center, University of Alabama, Birmingham, AL, 35294
| | - Sushant Bhatnagar
- Heersink School of Medicine, Division of Endocrinology, Diabetes, & Metabolism, Comprehensive Diabetes Center, University of Alabama, Birmingham, AL, 35294
| |
Collapse
|
5
|
Chen J, Wang Z, Wang T, Cheng J, Zhuang R, Wang W. SNAP23 decreases insulin secretion by competitively inhibiting the interaction between SNAP25 and STX1A. Biosci Rep 2023; 43:BSR20222594. [PMID: 37057886 PMCID: PMC10154458 DOI: 10.1042/bsr20222594] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 04/08/2023] [Accepted: 04/12/2023] [Indexed: 04/15/2023] Open
Abstract
SNAP25 is a core protein of the SNARE complex, which mediates stimulus-dependent secretion of insulin from the pancreatic β cells. SNAP23 is a SNAP25 homolog, however, the functional role of SNAP23 in the exocytic secretion of insulin is not known. Therefore, in the present study, we investigated the functional role of SNAP23 in the insulin secretory pathway. Our results demonstrated that over-expression of SNAP23 inhibited the secretion of insulin from the INS-1 cells. Conversely, SNAP23 depletion increased insulin secretion. Mechanistically, overexpression of SNAP23 decreased SNARE complex formation by blocking the binding of SNAP25 to STX1A. The full-length SNAP23 protein with the N-terminal and C-terminal SNARE binding domains was required for competition. Moreover, SNAP23 serine 95 phosphorylation plays a crucial function in insulin secretion by enhancing the interaction between SNAP23 and STX1A. The present study presents a new pathway regulating insulin secretion. Therefore, SNAP23 may be a potential therapeutic target for diabetes mellitus.
Collapse
Affiliation(s)
- Jun Chen
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361104, China
| | - Ziyan Wang
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361104, China
| | - Tuanlao Wang
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361104, China
| | - Jidong Cheng
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361104, China
| | - Ruijuan Zhuang
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361104, China
| | - Wei Wang
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361104, China
| |
Collapse
|
6
|
Buzzatto MV, Berberián MV, Di Bartolo AL, Masone D, Tomes CN. α-Synuclein is required for sperm exocytosis at a post-fusion stage. Front Cell Dev Biol 2023; 11:1125988. [PMID: 37287458 PMCID: PMC10242118 DOI: 10.3389/fcell.2023.1125988] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/09/2023] [Indexed: 06/09/2023] Open
Abstract
The sperm acrosome is a large dense-core granule whose contents are secreted by regulated exocytosis at fertilization through the opening of numerous fusion pores between the acrosomal and plasma membranes. In other cells, the nascent pore generated when the membrane surrounding a secretory vesicle fuses with the plasma membrane may have different fates. In sperm, pore dilation leads to the vesiculation and release of these membranes, together with the granule contents. α-Synuclein is a small cytosolic protein claimed to exhibit different roles in exocytic pathways in neurons and neuroendocrine cells. Here, we scrutinized its function in human sperm. Western blot revealed the presence of α-synuclein and indirect immunofluorescence its localization to the acrosomal domain of human sperm. Despite its small size, the protein was retained following permeabilization of the plasma membrane with streptolysin O. α-Synuclein was required for acrosomal release, as demonstrated by the inability of an inducer to elicit exocytosis when permeabilized human sperm were loaded with inhibitory antibodies to human α-synuclein. The antibodies halted calcium-induced secretion when introduced after the acrosome docked to the cell membrane. Two functional assays, fluorescence and transmission electron microscopies revealed that the stabilization of open fusion pores was responsible for the secretion blockage. Interestingly, synaptobrevin was insensitive to neurotoxin cleavage at this point, an indication of its engagement in cis SNARE complexes. The very existence of such complexes during AE reflects a new paradigm. Recombinant α-synuclein rescued the inhibitory effects of the anti-α-synuclein antibodies and of a chimeric Rab3A-22A protein that also inhibits AE after fusion pore opening. We applied restrained molecular dynamics simulations to compare the energy cost of expanding a nascent fusion pore between two model membranes and found it higher in the absence than in the presence of α-synuclein. Hence, our results suggest that α-synuclein is essential for expanding fusion pores.
Collapse
Affiliation(s)
- Micaela Vanina Buzzatto
- Instituto de Histología y Embriología de Mendoza (IHEM)-CONICET-Universidad Nacional de Cuyo, Mendoza, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María Victoria Berberián
- Instituto de Histología y Embriología de Mendoza (IHEM)-CONICET-Universidad Nacional de Cuyo, Mendoza, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
- Instituto de Ciencias Básicas (ICB)-CONICET-Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Ary Lautaro Di Bartolo
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Diego Masone
- Instituto de Histología y Embriología de Mendoza (IHEM)-CONICET-Universidad Nacional de Cuyo, Mendoza, Argentina
- Facultad de Ingeniería, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Claudia Nora Tomes
- Instituto de Histología y Embriología de Mendoza (IHEM)-CONICET-Universidad Nacional de Cuyo, Mendoza, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
7
|
Kang F, Xie L, Qin T, Miao Y, Kang Y, Takahashi T, Liang T, Xie H, Gaisano HY. Plasma membrane flipping of Syntaxin-2 regulates its inhibitory action on insulin granule exocytosis. Nat Commun 2022; 13:6512. [PMID: 36316316 PMCID: PMC9622911 DOI: 10.1038/s41467-022-33986-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 10/05/2022] [Indexed: 11/07/2022] Open
Abstract
Enhancing pancreatic β-cell secretion is a primary therapeutic target for type-2 diabetes (T2D). Syntaxin-2 (Stx2) has just been identified to be an inhibitory SNARE for insulin granule exocytosis, holding potential as a treatment for T2D, yet its molecular underpinnings remain unclear. We show that excessive Stx2 recruitment to raft-like granule docking sites at higher binding affinity than pro-fusion syntaxin-1A effectively competes for and inhibits fusogenic SNARE machineries. Depletion of Stx2 in human β-cells improves insulin secretion by enhancing trans-SNARE complex assembly and cis-SNARE disassembly. Using a genetically-encoded reporter, glucose stimulation is shown to induce Stx2 flipping across the plasma membrane, which relieves its suppression of cytoplasmic fusogenic SNARE complexes to promote insulin secretion. Targeting the flipping efficiency of Stx2 profoundly modulates secretion, which could restore the impaired insulin secretion in diabetes. Here, we show that Stx2 acts to assist this precise tuning of insulin secretion in β-cells, including in diabetes.
Collapse
Affiliation(s)
- Fei Kang
- grid.17063.330000 0001 2157 2938Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada ,grid.231844.80000 0004 0474 0428Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4 Canada
| | - Li Xie
- grid.17063.330000 0001 2157 2938Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Tairan Qin
- grid.17063.330000 0001 2157 2938Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Yifan Miao
- grid.17063.330000 0001 2157 2938Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Youhou Kang
- grid.17063.330000 0001 2157 2938Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Toshimasa Takahashi
- grid.17063.330000 0001 2157 2938Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Tao Liang
- grid.17063.330000 0001 2157 2938Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada ,grid.231844.80000 0004 0474 0428Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4 Canada
| | - Huanli Xie
- grid.17063.330000 0001 2157 2938Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Herbert Y. Gaisano
- grid.17063.330000 0001 2157 2938Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada ,grid.231844.80000 0004 0474 0428Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4 Canada
| |
Collapse
|
8
|
Fan F, Wu Y, Hara M, Rizk A, Ji C, Nerad D, Tamarina N, Lou X. Dynamin deficiency causes insulin secretion failure and hyperglycemia. Proc Natl Acad Sci U S A 2021; 118:e2021764118. [PMID: 34362840 PMCID: PMC8364113 DOI: 10.1073/pnas.2021764118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pancreatic β cells operate with a high rate of membrane recycling for insulin secretion, yet endocytosis in these cells is not fully understood. We investigate this process in mature mouse β cells by genetically deleting dynamin GTPase, the membrane fission machinery essential for clathrin-mediated endocytosis. Unexpectedly, the mice lacking all three dynamin genes (DNM1, DNM2, DNM3) in their β cells are viable, and their β cells still contain numerous insulin granules. Endocytosis in these β cells is severely impaired, resulting in abnormal endocytic intermediates on the plasma membrane. Although insulin granules are abundant, their release upon glucose stimulation is blunted in both the first and second phases, leading to hyperglycemia and glucose intolerance in mice. Dynamin triple deletion impairs insulin granule exocytosis and decreases intracellular Ca2+ responses and granule docking. The docking defect is correlated with reduced expression of Munc13-1 and RIM1 and reorganization of cortical F-actin in β cells. Collectively, these findings uncover the role of dynamin in dense-core vesicle endocytosis and secretory capacity. Insulin secretion deficiency in the absence of dynamin-mediated endocytosis highlights the risk of impaired membrane trafficking in endocrine failure and diabetes pathogenesis.
Collapse
Affiliation(s)
- Fan Fan
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Yumei Wu
- HHMI, Yale University School of Medicine, New Haven, CT 06510
- Departments of Neuroscience and Cell Biology, Program in Cellular Neuroscience, Neurodegeneration and Repair, Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT 06510
| | - Manami Hara
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Adam Rizk
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Chen Ji
- Synapses and Circuits section, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892
| | - Dan Nerad
- Emergency Medicine, Carl R. Darnall Army Medical Center, Fort Hood, TX 76544
| | - Natalia Tamarina
- Department of Medicine, The Kovler Diabetes Center, University of Chicago, Chicago, IL 60637
| | - Xuelin Lou
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226;
| |
Collapse
|
9
|
Gerber KM, Whitticar NB, Rochester DR, Corbin KL, Koch WJ, Nunemaker CS. The Capacity to Secrete Insulin Is Dose-Dependent to Extremely High Glucose Concentrations: A Key Role for Adenylyl Cyclase. Metabolites 2021; 11:metabo11060401. [PMID: 34205432 PMCID: PMC8235240 DOI: 10.3390/metabo11060401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 12/12/2022] Open
Abstract
Insulin secretion is widely thought to be maximally stimulated in glucose concentrations of 16.7-to-30 mM (300-to-540 mg/dL). However, insulin secretion is seldom tested in hyperglycemia exceeding these levels despite the Guinness World Record being 147.6 mM (2656 mg/dL). We investigated how islets respond to 1-h exposure to glucose approaching this record. Insulin secretion from human islets at 12 mM glucose intervals dose-dependently increased until at least 72 mM glucose. Murine islets in 84 mM glucose secreted nearly double the insulin as in 24 mM (p < 0.001). Intracellular calcium was maximally stimulated in 24 mM glucose despite a further doubling of insulin secretion in higher glucose, implying that insulin secretion above 24 mM occurs through amplifying pathway(s). Increased osmolarity of 425-mOsm had no effect on insulin secretion (1-h exposure) or viability (48-h exposure) in murine islets. Murine islets in 24 mM glucose treated with a glucokinase activator secreted as much insulin as islets in 84 mM glucose, indicating that glycolytic capacity exists above 24 mM. Using an incretin mimetic and an adenylyl cyclase activator in 24 mM glucose enhanced insulin secretion above that observed in 84 mM glucose while adenylyl cyclase inhibitor reduced stimulatory effects. These results highlight the underestimated ability of islets to secrete insulin proportionally to extreme hyperglycemia through adenylyl cyclase activity.
Collapse
Affiliation(s)
- Katherine M. Gerber
- Translational Health, Honors Tutorial College, Ohio University, Athens, OH 45701, USA;
| | - Nicholas B. Whitticar
- Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; (N.B.W.); (D.R.R.); (K.L.C.); (W.J.K.)
- Translational Biomedical Sciences Program, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Daniel R. Rochester
- Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; (N.B.W.); (D.R.R.); (K.L.C.); (W.J.K.)
| | - Kathryn L. Corbin
- Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; (N.B.W.); (D.R.R.); (K.L.C.); (W.J.K.)
| | - William J. Koch
- Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; (N.B.W.); (D.R.R.); (K.L.C.); (W.J.K.)
- Translational Biomedical Sciences Program, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Craig S. Nunemaker
- Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; (N.B.W.); (D.R.R.); (K.L.C.); (W.J.K.)
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Correspondence: ; Tel.: +740-593-2387
| |
Collapse
|
10
|
Isolation and Proteomics of the Insulin Secretory Granule. Metabolites 2021; 11:metabo11050288. [PMID: 33946444 PMCID: PMC8147143 DOI: 10.3390/metabo11050288] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 12/21/2022] Open
Abstract
Insulin, a vital hormone for glucose homeostasis is produced by pancreatic beta-cells and when secreted, stimulates the uptake and storage of glucose from the blood. In the pancreas, insulin is stored in vesicles termed insulin secretory granules (ISGs). In Type 2 diabetes (T2D), defects in insulin action results in peripheral insulin resistance and beta-cell compensation, ultimately leading to dysfunctional ISG production and secretion. ISGs are functionally dynamic and many proteins present either on the membrane or in the lumen of the ISG may modulate and affect different stages of ISG trafficking and secretion. Previously, studies have identified few ISG proteins and more recently, proteomics analyses of purified ISGs have uncovered potential novel ISG proteins. This review summarizes the proteins identified in the current ISG proteomes from rat insulinoma INS-1 and INS-1E cell lines. Here, we also discuss techniques of ISG isolation and purification, its challenges and potential future directions.
Collapse
|
11
|
Chatterjee Bhowmick D, Ahn M, Oh E, Veluthakal R, Thurmond DC. Conventional and Unconventional Mechanisms by which Exocytosis Proteins Oversee β-cell Function and Protection. Int J Mol Sci 2021; 22:1833. [PMID: 33673206 PMCID: PMC7918544 DOI: 10.3390/ijms22041833] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/02/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetes (T2D) is one of the prominent causes of morbidity and mortality in the United States and beyond, reaching global pandemic proportions. One hallmark of T2D is dysfunctional glucose-stimulated insulin secretion from the pancreatic β-cell. Insulin is secreted via the recruitment of insulin secretory granules to the plasma membrane, where the soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) and SNARE regulators work together to dock the secretory granules and release insulin into the circulation. SNARE proteins and their regulators include the Syntaxins, SNAPs, Sec1/Munc18, VAMPs, and double C2-domain proteins. Recent studies using genomics, proteomics, and biochemical approaches have linked deficiencies of exocytosis proteins with the onset and progression of T2D. Promising results are also emerging wherein restoration or enhancement of certain exocytosis proteins to β-cells improves whole-body glucose homeostasis, enhances β-cell function, and surprisingly, protection of β-cell mass. Intriguingly, overexpression and knockout studies have revealed novel functions of certain exocytosis proteins, like Syntaxin 4, suggesting that exocytosis proteins can impact a variety of pathways, including inflammatory signaling and aging. In this review, we present the conventional and unconventional functions of β-cell exocytosis proteins in normal physiology and T2D and describe how these insights might improve clinical care for T2D.
Collapse
Affiliation(s)
| | | | | | | | - Debbie C. Thurmond
- Department of Molecular and Cellular Endocrinology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; (D.C.B.); (M.A.); (E.O.); (R.V.)
| |
Collapse
|
12
|
Balboa D, Iworima DG, Kieffer TJ. Human Pluripotent Stem Cells to Model Islet Defects in Diabetes. Front Endocrinol (Lausanne) 2021; 12:642152. [PMID: 33828531 PMCID: PMC8020750 DOI: 10.3389/fendo.2021.642152] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/03/2021] [Indexed: 12/17/2022] Open
Abstract
Diabetes mellitus is characterized by elevated levels of blood glucose and is ultimately caused by insufficient insulin production from pancreatic beta cells. Different research models have been utilized to unravel the molecular mechanisms leading to the onset of diabetes. The generation of pancreatic endocrine cells from human pluripotent stem cells constitutes an approach to study genetic defects leading to impaired beta cell development and function. Here, we review the recent progress in generating and characterizing functional stem cell-derived beta cells. We summarize the diabetes disease modeling possibilities that stem cells offer and the challenges that lie ahead to further improve these models.
Collapse
Affiliation(s)
- Diego Balboa
- Regulatory Genomics and Diabetes, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- *Correspondence: Diego Balboa,
| | - Diepiriye G. Iworima
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Timothy J. Kieffer
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
13
|
Wilhelmi I, Grunwald S, Gimber N, Popp O, Dittmar G, Arumughan A, Wanker EE, Laeger T, Schmoranzer J, Daumke O, Schürmann A. The ARFRP1-dependent Golgi scaffolding protein GOPC is required for insulin secretion from pancreatic β-cells. Mol Metab 2020; 45:101151. [PMID: 33359402 PMCID: PMC7811047 DOI: 10.1016/j.molmet.2020.101151] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 12/13/2022] Open
Abstract
Objective Hormone secretion from metabolically active tissues, such as pancreatic islets, is governed by specific and highly regulated signaling pathways. Defects in insulin secretion are among the major causes of diabetes. The molecular mechanisms underlying regulated insulin secretion are, however, not yet completely understood. In this work, we studied the role of the GTPase ARFRP1 on insulin secretion from pancreatic β-cells. Methods A β-cell-specific Arfrp1 knockout mouse was phenotypically characterized. Pulldown experiments and mass spectrometry analysis were employed to screen for new ARFRP1-interacting proteins. Co-immunoprecipitation assays as well as super-resolution microscopy were applied for validation. Results The GTPase ARFRP1 interacts with the Golgi-associated PDZ and coiled-coil motif-containing protein (GOPC). Both proteins are co-localized at the trans-Golgi network and regulate the first and second phase of insulin secretion by controlling the plasma membrane localization of the SNARE protein SNAP25. Downregulation of both GOPC and ARFRP1 in Min6 cells interferes with the plasma membrane localization of SNAP25 and enhances its degradation, thereby impairing glucose-stimulated insulin release from β-cells. In turn, overexpression of SNAP25 as well as GOPC restores insulin secretion in islets from β-cell-specific Arfrp1 knockout mice. Conclusion Our results identify a hitherto unrecognized pathway required for insulin secretion at the level of trans-Golgi sorting. β-cell specific deletion of the trans-Golgi residing small GTPase ARFRP1 leads to elevated blood glucose levels in mice. GOPC is a newly identified ARFRP1 dependent scaffolding protein. ARFRP1 and GOPC are required for glucose-stimulated insulin secretion from pancreatic β-cells.
Collapse
Affiliation(s)
- Ilka Wilhelmi
- German Institute of Human Nutrition (DIfE) Potsdam-Rehbruecke, Germany; German Center for Diabetes Research (DZD) Munich Neuherberg, Germany
| | - Stephan Grunwald
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Germany; Institute of Chemistry and Biochemistry, Freie Universität Berlin, Germany
| | - Niclas Gimber
- Advanced Medical Bioimaging Core Facility - AMBIO, Charité-Universitätsmedizin Berlin, Germany
| | - Oliver Popp
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Germany
| | - Gunnar Dittmar
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Germany
| | - Anup Arumughan
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) Berlin, Germany
| | - Erich E Wanker
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) Berlin, Germany
| | - Thomas Laeger
- German Institute of Human Nutrition (DIfE) Potsdam-Rehbruecke, Germany; German Center for Diabetes Research (DZD) Munich Neuherberg, Germany
| | - Jan Schmoranzer
- Advanced Medical Bioimaging Core Facility - AMBIO, Charité-Universitätsmedizin Berlin, Germany
| | - Oliver Daumke
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Germany; Institute of Chemistry and Biochemistry, Freie Universität Berlin, Germany
| | - Annette Schürmann
- German Institute of Human Nutrition (DIfE) Potsdam-Rehbruecke, Germany; German Center for Diabetes Research (DZD) Munich Neuherberg, Germany; University of Potsdam, Institute of Nutritional Sciences, Nuthetal, Germany; Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, the Brandenburg Medical School Theodor Fontane and the University of Potsdam, Germany.
| |
Collapse
|
14
|
Gheibi S, Ghasemi A. Insulin secretion: The nitric oxide controversy. EXCLI JOURNAL 2020; 19:1227-1245. [PMID: 33088259 PMCID: PMC7573190 DOI: 10.17179/excli2020-2711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 08/31/2020] [Indexed: 12/14/2022]
Abstract
Nitric oxide (NO) is a gas that serves as a ubiquitous signaling molecule participating in physiological activities of various organ systems. Nitric oxide is produced in the endocrine pancreas and contributes to synthesis and secretion of insulin. The potential role of NO in insulin secretion is disputable - both stimulatory and inhibitory effects have been reported. Available data indicate that effects of NO critically depend on its concentration. Different isoforms of NO synthase (NOS) control this and have the potential to decrease or increase insulin secretion. In this review, the role of NO in insulin secretion as well as the possible reasons for discrepant findings are discussed. A better understanding of the role of NO system in the regulation of insulin secretion may facilitate the development of new therapeutic strategies in the management of diabetes.
Collapse
Affiliation(s)
- Sevda Gheibi
- Department of Clinical Sciences in Malmö, Unit of Molecular Metabolism, Lund University Diabetes Centre, Clinical Research Center, Malmö University Hospital, Lund University, Malmö, Sweden
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Yau B, Hays L, Liang C, Laybutt DR, Thomas HE, Gunton JE, Williams L, Hawthorne WJ, Thorn P, Rhodes CJ, Kebede MA. A fluorescent timer reporter enables sorting of insulin secretory granules by age. J Biol Chem 2020; 295:8901-8911. [PMID: 32341128 PMCID: PMC7335792 DOI: 10.1074/jbc.ra120.012432] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 03/21/2020] [Indexed: 01/03/2023] Open
Abstract
Within the pancreatic β-cells, insulin secretory granules (SGs) exist in functionally distinct pools, displaying variations in motility as well as docking and fusion capability. Current therapies that increase insulin secretion do not consider the existence of these distinct SG pools. Accordingly, these approaches are effective only for a short period, with a worsening of glycemia associated with continued decline in β-cell function. Insulin granule age is underappreciated as a determinant for why an insulin granule is selected for secretion and may explain why newly synthesized insulin is preferentially secreted from β-cells. Here, using a novel fluorescent timer protein, we aimed to investigate the preferential secretion model of insulin secretion and identify how granule aging is affected by variation in the β-cell environment, such as hyperglycemia. We demonstrate the use of a fluorescent timer construct, syncollin-dsRedE5TIMER, which changes its fluorescence from green to red over 18 h, in both microscopy and fluorescence-assisted organelle-sorting techniques. We confirm that the SG-targeting construct localizes to insulin granules in β-cells and does not interfere with normal insulin SG behavior. We visualize insulin SG aging behavior in MIN6 and INS1 β-cell lines and in primary C57BL/6J mouse and nondiabetic human islet cells. Finally, we separated young and old insulin SGs, revealing that preferential secretion of younger granules occurs in glucose-stimulated insulin secretion. We also show that SG population age is modulated by the β-cell environment in vivo in the db/db mouse islets and ex vivo in C57BL/6J islets exposed to different glucose environments.
Collapse
Affiliation(s)
- Belinda Yau
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia; School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Sydney, New South Wales, Australia
| | - Lori Hays
- STEM-Department of Biology, Edmonds Community College, Lynnwood, Washington, USA
| | - Cassandra Liang
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - D Ross Laybutt
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia; St. Vincent's Clinical School, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Helen E Thomas
- St. Vincent's Institute, Fitzroy, Victoria, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
| | - Jenny E Gunton
- Faculty of Medicine and Health, the University of Sydney, Sydney, New South Wales, Australia; The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Lindy Williams
- Faculty of Medicine and Health, the University of Sydney, Sydney, New South Wales, Australia; National Pancreas and Islet Transplant Unit (NPITU), Westmead Hospital, Sydney, New South Wales, Australia
| | - Wayne J Hawthorne
- Faculty of Medicine and Health, the University of Sydney, Sydney, New South Wales, Australia; National Pancreas and Islet Transplant Unit (NPITU), Westmead Hospital, Sydney, New South Wales, Australia
| | - Peter Thorn
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia; Discipline of Physiology, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Christopher J Rhodes
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca Ltd, Gaithersburg, Maryland, USA; Pacific Northwest Research Institute, Seattle, Washington, USA
| | - Melkam A Kebede
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia; School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
16
|
Berdous D, Berney X, Sanchez-Archidona AR, Jan M, Roujeau C, Lopez-Mejia IC, Mynatt R, Thorens B. A genetic screen identifies Crat as a regulator of pancreatic beta-cell insulin secretion. Mol Metab 2020; 37:100993. [PMID: 32298772 PMCID: PMC7225740 DOI: 10.1016/j.molmet.2020.100993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/02/2020] [Accepted: 04/02/2020] [Indexed: 11/28/2022] Open
Abstract
Objectives Glucose-stimulated insulin secretion is a critical function in the regulation of glucose homeostasis, and its deregulation is associated with the development of type 2 diabetes. Here, we performed a genetic screen using islets isolated from the BXD panel of advanced recombinant inbred (RI) lines of mice to search for novel regulators of insulin production and secretion. Methods Pancreatic islets were isolated from 36 RI BXD lines and insulin secretion was measured following exposure to 2.8 or 16.7 mM glucose with or without exendin-4. Islets from the same RI lines were used for RNA extraction and transcript profiling. Quantitative trait loci (QTL) mapping was performed for each secretion condition and combined with transcriptome data to prioritize candidate regulatory genes within the identified QTL regions. Functional studies were performed by mRNA silencing or overexpression in MIN6B1 cells and by studying mice and islets with beta-cell-specific gene inactivation. Results Insulin secretion under the 16.7 mM glucose plus exendin-4 condition was mapped significantly to a chromosome 2 QTL. Within this QTL, RNA-Seq data prioritized Crat (carnitine O-acetyl transferase) as a strong candidate regulator of the insulin secretion trait. Silencing Crat expression in MIN6B1 cells reduced insulin content and insulin secretion by ∼30%. Conversely, Crat overexpression enhanced insulin content and secretion by ∼30%. When islets from mice with beta-cell-specific Crat inactivation were exposed to high glucose, they displayed a 30% reduction of insulin content as compared to control islets. We further showed that decreased Crat expression in both MIN6B1 cells and pancreatic islets reduced the oxygen consumption rate in a glucose concentration-dependent manner. Conclusions We identified Crat as a regulator of insulin secretion whose action is mediated by an effect on total cellular insulin content; this effect also depends on the genetic background of the RI mouse lines. These data also show that in the presence of the stimulatory conditions used the insulin secretion rate is directly related to the insulin content. A QTL analysis in BXD mice identifies Crat as a regulator of insulin secretion. Crat regulates insulin content in MIN6B1 cells and pancreatic islets. Crat regulates glucose oxidation in MIN6B1 cells and pancreatic islets. Crat links glucose metabolism to the control of beta-cell insulin content. Insulin content limits insulin secretion in response to high glucose and exendin-4 level.
Collapse
Affiliation(s)
- Dassine Berdous
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| | - Xavier Berney
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| | - Ana Rodriguez Sanchez-Archidona
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland; Vital-IT, Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland.
| | - Maxime Jan
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| | - Clara Roujeau
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| | - Isabel C Lopez-Mejia
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| | - Randall Mynatt
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA.
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| |
Collapse
|
17
|
Abstract
Glucose-induced (physiological) insulin secretion from the islet β-cell involves interplay between cationic (i.e., changes in intracellular calcium) and metabolic (i.e., generation of hydrophobic and hydrophilic second messengers) events. A large body of evidence affirms support for novel regulation, by G proteins, of specific intracellular signaling events, including actin cytoskeletal remodeling, transport of insulin-containing granules to the plasma membrane for fusion, and secretion of insulin into the circulation. This article highlights the following aspects of GPCR-G protein biology of the islet. First, it overviews our current understanding of the identity of a wide variety of G protein regulators and their modulatory roles in GPCR-G protein-effector coupling, which is requisite for optimal β-cell function under physiological conditions. Second, it describes evidence in support of novel, noncanonical, GPCR-independent mechanisms of activation of G proteins in the islet. Third, it highlights the evidence indicating that abnormalities in G protein function lead to islet β-cell dysregulation and demise under the duress of metabolic stress and diabetes. Fourth, it summarizes observations of potential beneficial effects of GPCR agonists in preventing/halting metabolic defects in the islet β-cell under various pathological conditions (e.g., metabolic stress and inflammation). Lastly, it identifies knowledge gaps and potential avenues for future research in this evolving field of translational islet biology. Published 2020. Compr Physiol 10:453-490, 2020.
Collapse
Affiliation(s)
- Anjaneyulu Kowluru
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Center for Translational Research in Diabetes, Biomedical Research Service, John D. Dingell VA Medical Center, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
18
|
Thurmond DC, Gaisano HY. Recent Insights into Beta-cell Exocytosis in Type 2 Diabetes. J Mol Biol 2020; 432:1310-1325. [PMID: 31863749 PMCID: PMC8061716 DOI: 10.1016/j.jmb.2019.12.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 11/26/2019] [Accepted: 12/05/2019] [Indexed: 01/26/2023]
Abstract
As one of the leading causes of morbidity and mortality worldwide, diabetes affects an estimated 422 million adults, and it is expected to continue expanding such that by 2050, 30% of the U.S. population will become diabetic within their lifetime. Out of the estimated 422 million people currently afflicted with diabetes worldwide, about 5% have type 1 diabetes (T1D), while the remaining ~95% of diabetics have type 2 diabetes (T2D). Type 1 diabetes results from the autoimmune-mediated destruction of functional β-cell mass, whereas T2D results from combinatorial defects in functional β-cell mass plus peripheral glucose uptake. Both types of diabetes are now believed to be preceded by β-cell dysfunction. T2D is increasingly associated with numerous reports of deficiencies in the exocytosis proteins that regulate insulin release from β-cells, specifically the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins. SNARE protein's functionality is further regulated by a variety of accessory factors such as Sec1/Munc18 (SM), double C2-domain proteins (DOC2), and additional interacting proteins at the cell surface that influence the fidelity of insulin release. As new evidence emerges about the detailed mechanisms of exocytosis, new questions and controversies have come to light. This emerging information is also contributing to dialogue in the islet biology field focused on how to correct the defects in insulin exocytosis. Herein we present a balanced review of the role of exocytosis proteins in T2D, with thoughts on novel strategies to protect functional β-cell mass.
Collapse
Affiliation(s)
- Debbie C Thurmond
- Department of Molecular and Cellular Endocrinology, Beckman Research Institute of City of Hope, CA, USA.
| | | |
Collapse
|
19
|
Liang T, Qin T, Kang F, Kang Y, Xie L, Zhu D, Dolai S, Greitzer-Antes D, Baker RK, Feng D, Tuduri E, Ostenson CG, Kieffer TJ, Banks K, Pessin JE, Gaisano HY. SNAP23 depletion enables more SNAP25/calcium channel excitosome formation to increase insulin exocytosis in type 2 diabetes. JCI Insight 2020; 5:129694. [PMID: 32051343 PMCID: PMC7098801 DOI: 10.1172/jci.insight.129694] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 01/15/2020] [Indexed: 01/05/2023] Open
Abstract
SNAP23 is the ubiquitous SNAP25 isoform that mediates secretion in non-neuronal cells, similar to SNAP25 in neurons. However, some secretory cells like pancreatic islet β cells contain an abundance of both SNAP25 and SNAP23, where SNAP23 is believed to play a redundant role to SNAP25. We show that SNAP23, when depleted in mouse β cells in vivo and human β cells (normal and type 2 diabetes [T2D] patients) in vitro, paradoxically increased biphasic glucose-stimulated insulin secretion corresponding to increased exocytosis of predocked and newcomer insulin granules. Such effects on T2D Goto-Kakizaki rats improved glucose homeostasis that was superior to conventional treatment with sulfonylurea glybenclamide. SNAP23, although fusion competent in slower secretory cells, in the context of β cells acts as a weak partial fusion agonist or inhibitory SNARE. Here, SNAP23 depletion promotes SNAP25 to bind calcium channels more quickly and longer where granule fusion occurs to increase exocytosis efficiency. β Cell SNAP23 antagonism is a strategy to treat diabetes.
Collapse
Affiliation(s)
- Tao Liang
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Tairan Qin
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Fei Kang
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Youhou Kang
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Li Xie
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Dan Zhu
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Subhankar Dolai
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Dafna Greitzer-Antes
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Robert K. Baker
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Daorong Feng
- Michael F. Price Center for Genetic and Translational Medicine, Department of Medicine and Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Eva Tuduri
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Claes-Goran Ostenson
- Department of Molecular Medicine and,Department of Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Timothy J. Kieffer
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kate Banks
- Division of Comparative Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jeffrey E. Pessin
- Michael F. Price Center for Genetic and Translational Medicine, Department of Medicine and Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Herbert Y. Gaisano
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Ruete MC, Zarelli VEP, Masone D, de Paola M, Bustos DM, Tomes CN. A connection between reversible tyrosine phosphorylation and SNARE complex disassembly activity of N-ethylmaleimide-sensitive factor unveiled by the phosphomimetic mutant N-ethylmaleimide-sensitive factor-Y83E. ACTA ACUST UNITED AC 2019; 25:344-358. [DOI: 10.1093/molehr/gaz031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/16/2019] [Accepted: 06/03/2019] [Indexed: 12/26/2022]
Abstract
Abstract
N-ethylmaleimide-sensitive factor (NSF) disassembles fusion-incompetent cis soluble-NSF attachment protein receptor (SNARE) complexes making monomeric SNAREs available for subsequent trans pairing and fusion. In most cells the activity of NSF is constitutive, but in Jurkat cells and sperm it is repressed by tyrosine phosphorylation; the phosphomimetic mutant NSF–Y83E inhibits secretion in the former. The questions addressed here are if and how the NSF mutant influences the configuration of the SNARE complex. Our model is human sperm, where the initiation of exocytosis (acrosome reaction (AR)) de-represses the activity of NSF through protein tyrosine phosphatase 1B (PTP1B)-mediated dephosphorylation. We developed a fluorescence microscopy-based method to show that capacitation increased, and challenging with an AR inducer decreased, the number of cells with tyrosine-phosphorylated PTP1B substrates in the acrosomal domain. Results from bioinformatic and biochemical approaches using purified recombinant proteins revealed that NSF–Y83E bound PTP1B and thereupon inhibited its catalytic activity. Mutant NSF introduced into streptolysin O-permeabilized sperm impaired cis SNARE complex disassembly, blocking the AR; subsequent addition of PTP1B rescued exocytosis. We propose that NSF–Y83E prevents endogenous PTP1B from dephosphorylating sperm NSF, thus maintaining NSF’s activity in a repressed mode and the SNARE complex unable to dissociate. The contribution of this paper to the sperm biology field is the detection of PTP1B substrates, one of them likely being NSF, whose tyrosine phosphorylation status varies during capacitation and the AR. The contribution of this paper to the membrane traffic field is to have generated direct evidence that explains the dominant-negative role of the phosphomimetic mutant NSF–Y83E.
Collapse
Affiliation(s)
- María Celeste Ruete
- Instituto de Histología y Embriología de Mendoza Dr Mario H. Burgos–CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Valeria Eugenia Paola Zarelli
- Instituto de Histología y Embriología de Mendoza Dr Mario H. Burgos–CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Diego Masone
- Instituto de Histología y Embriología de Mendoza Dr Mario H. Burgos–CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina
- Facultad de Ingeniería, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Matilde de Paola
- Instituto de Histología y Embriología de Mendoza Dr Mario H. Burgos–CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
- Instituto de Medicina y Biología Experimental de Cuyo–Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Diego Martín Bustos
- Instituto de Histología y Embriología de Mendoza Dr Mario H. Burgos–CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Claudia Nora Tomes
- Instituto de Histología y Embriología de Mendoza Dr Mario H. Burgos–CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
21
|
Dufurrena Q, Bäck N, Mains R, Hodgson L, Tanowitz H, Mandela P, Eipper B, Kuliawat R. Kalirin/Trio Rho GDP/GTP exchange factors regulate proinsulin and insulin secretion. J Mol Endocrinol 2018; 62:JME-18-0048.R2. [PMID: 30407917 PMCID: PMC6494717 DOI: 10.1530/jme-18-0048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 11/05/2018] [Indexed: 12/31/2022]
Abstract
Key features for progression to pancreatic β-cell failure and disease are loss of glucose responsiveness and an increased ratio of secreted proinsulin to insulin. Proinsulin and insulin are stored in secretory granules (SGs) and the fine-tuning of hormone output requires signal mediated recruitment of select SG populations according to intracellular location and age. The GTPase Rac1 coordinates multiple signaling pathways that specify SG release and Rac1 activity is controlled in part by GDP/GTP exchange factors (GEFs). To explore the function of two large multidomain GEFs, Kalirin and Trio in β-cells, we manipulated their Rac1-specific GEF1 domain activity by using small molecule inhibitors and by genetically ablating Kalirin. We examined age related secretory granule behavior employing radiolabeling protocols. Loss of Kalirin/Trio function attenuated radioactive proinsulin release by reducing constitutive-like secretion and exocytosis of 2-hour old granules. At later chase times or at steady state, Kalirin/Trio manipulations decreased glucose stimulated insulin output. Finally, use of a Rac1 FRET biosensor with cultured β-cell lines, demonstrated that Kalirin/Trio GEF1 activity was required for normal rearrangement of Rac1 to the plasma membrane in response to glucose. Rac1 activation can be evoked by both glucose metabolism and signaling through the incretin glucagon-like peptide 1 (GLP-1) receptor. GLP-1 addition restored Rac1 localization/activity and insulin secretion in the absence of Kalirin, thereby assigning Kalirin's participation to stimulatory glucose signaling.
Collapse
Affiliation(s)
- Quinn Dufurrena
- Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY
| | - Nils Bäck
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Richard Mains
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT
| | - Louis Hodgson
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Herbert Tanowitz
- Departments of Pathology, Medicine, Albert Einstein College of Medicine, Bronx, NY
| | | | - Betty Eipper
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT
| | - Regina Kuliawat
- Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
22
|
Greitzer-Antes D, Xie L, Qin T, Xie H, Zhu D, Dolai S, Liang T, Kang F, Hardy AB, He Y, Kang Y, Gaisano HY. K v2.1 clusters on β-cell plasma membrane act as reservoirs that replenish pools of newcomer insulin granule through their interaction with syntaxin-3. J Biol Chem 2018; 293:6893-6904. [PMID: 29549124 DOI: 10.1074/jbc.ra118.002703] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 03/09/2018] [Indexed: 01/22/2023] Open
Abstract
The voltage-dependent K+ (Kv) channel Kv2.1 is a major delayed rectifier in many secretory cells, including pancreatic β cells. In addition, Kv2.1 has a direct role in exocytosis at an undefined step, involving SNARE proteins, that is independent of its ion-conducting pore function. Here, we elucidated the precise step in exocytosis. We previously reported that syntaxin-3 (Syn-3) is the key syntaxin that mediates exocytosis of newcomer secretory granules that spend minimal residence time on the plasma membrane before fusion. Using high-resolution total internal reflection fluorescence microscopy, we now show that Kv2.1 forms reservoir clusters on the β-cell plasma membrane and binds Syn-3 via its C-terminal C1b domain, which recruits newcomer insulin secretory granules into this large reservoir. Upon glucose stimulation, secretory granules were released from this reservoir to replenish the pool of newcomer secretory granules for subsequent fusion, occurring just adjacent to the plasma membrane Kv2.1 clusters. C1b deletion blocked the aforementioned Kv2.1-Syn-3-mediated events and reduced fusion of newcomer secretory granules. These insights have therapeutic implications, as Kv2.1 overexpression in type-2 diabetes rat islets restored biphasic insulin secretion.
Collapse
Affiliation(s)
- Dafna Greitzer-Antes
- From the Departments of Medicine and Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada and
| | - Li Xie
- From the Departments of Medicine and Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada and
| | - Tairan Qin
- From the Departments of Medicine and Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada and
| | - Huanli Xie
- From the Departments of Medicine and Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada and
| | - Dan Zhu
- From the Departments of Medicine and Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada and
| | - Subhankar Dolai
- From the Departments of Medicine and Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada and
| | - Tao Liang
- From the Departments of Medicine and Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada and
| | - Fei Kang
- From the Departments of Medicine and Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada and
| | - Alexandre B Hardy
- From the Departments of Medicine and Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada and
| | - Yan He
- the Department of Epidemiology and Health Statistics, School of Public Health and Family Medicine, Capital Medical University, Beijing 100050, China
| | - Youhou Kang
- From the Departments of Medicine and Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada and
| | - Herbert Y Gaisano
- From the Departments of Medicine and Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada and
| |
Collapse
|
23
|
Rorsman P, Ashcroft FM. Pancreatic β-Cell Electrical Activity and Insulin Secretion: Of Mice and Men. Physiol Rev 2018; 98:117-214. [PMID: 29212789 PMCID: PMC5866358 DOI: 10.1152/physrev.00008.2017] [Citation(s) in RCA: 514] [Impact Index Per Article: 73.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/30/2017] [Accepted: 06/18/2017] [Indexed: 12/14/2022] Open
Abstract
The pancreatic β-cell plays a key role in glucose homeostasis by secreting insulin, the only hormone capable of lowering the blood glucose concentration. Impaired insulin secretion results in the chronic hyperglycemia that characterizes type 2 diabetes (T2DM), which currently afflicts >450 million people worldwide. The healthy β-cell acts as a glucose sensor matching its output to the circulating glucose concentration. It does so via metabolically induced changes in electrical activity, which culminate in an increase in the cytoplasmic Ca2+ concentration and initiation of Ca2+-dependent exocytosis of insulin-containing secretory granules. Here, we review recent advances in our understanding of the β-cell transcriptome, electrical activity, and insulin exocytosis. We highlight salient differences between mouse and human β-cells, provide models of how the different ion channels contribute to their electrical activity and insulin secretion, and conclude by discussing how these processes become perturbed in T2DM.
Collapse
Affiliation(s)
- Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom; Department of Neuroscience and Physiology, Metabolic Research Unit, Göteborg, Sweden; and Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Frances M Ashcroft
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom; Department of Neuroscience and Physiology, Metabolic Research Unit, Göteborg, Sweden; and Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
24
|
Hastoy B, Clark A, Rorsman P, Lang J. Fusion pore in exocytosis: More than an exit gate? A β-cell perspective. Cell Calcium 2017; 68:45-61. [PMID: 29129207 DOI: 10.1016/j.ceca.2017.10.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/17/2017] [Accepted: 10/24/2017] [Indexed: 12/14/2022]
Abstract
Secretory vesicle exocytosis is a fundamental biological event and the process by which hormones (like insulin) are released into the blood. Considerable progress has been made in understanding this precisely orchestrated sequence of events from secretory vesicle docked at the cell membrane, hemifusion, to the opening of a membrane fusion pore. The exact biophysical and physiological regulation of these events implies a close interaction between membrane proteins and lipids in a confined space and constrained geometry to ensure appropriate delivery of cargo. We consider some of the still open questions such as the nature of the initiation of the fusion pore, the structure and the role of the Soluble N-ethylmaleimide-sensitive-factor Attachment protein REceptor (SNARE) transmembrane domains and their influence on the dynamics and regulation of exocytosis. We discuss how the membrane composition and protein-lipid interactions influence the likelihood of the nascent fusion pore forming. We relate these factors to the hypothesis that fusion pore expansion could be affected in type-2 diabetes via changes in disease-related gene transcription and alterations in the circulating lipid profile. Detailed characterisation of the dynamics of the fusion pore in vitro will contribute to understanding the larger issue of insulin secretory defects in diabetes.
Collapse
Affiliation(s)
- Benoit Hastoy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK.
| | - Anne Clark
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK; Metabolic Research, Institute of Neuroscience and Physiology, University of Goteborg, Medicinaregatan 11, S-41309 Göteborg, Sweden
| | - Jochen Lang
- Laboratoire de Chimie et Biologie des Membranes et Nano-objets (CBMN), CNRS UMR 5248, Université de Bordeaux, Allée de Geoffrey St Hilaire, 33600 Pessac, France.
| |
Collapse
|
25
|
Brüning D, Reckers K, Drain P, Rustenbeck I. Glucose but not KCl diminishes submembrane granule turnover in mouse beta-cells. J Mol Endocrinol 2017; 59:311-324. [PMID: 28765259 DOI: 10.1530/jme-17-0063] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 08/01/2017] [Indexed: 01/22/2023]
Abstract
KCl depolarization is widely used to mimic the depolarization during glucose-stimulated insulin secretion. Consequently, the insulin secretion elicited by KCl is often regarded as the equivalent of the first phase of glucose-induced insulin secretion. Here, the effects of both stimuli were compared by measuring the secretion of perifused mouse islets, the cytosolic Ca2+ concentration of single beta-cells and the mobility of submembrane insulin granules by TIRF microscopy of primary mouse beta-cells. Two cargo-directed granule labels were used namely insulin-EGFP and C-peptide-emGFP. The granule behaviour common to both was used to compare the effect of sequential stimulation with 40 mM KCl and 30 mM glucose and sequential stimulation with the same stimuli in reversed order. At the level of the cell secretory response, the sequential pulse protocol showed marked differences depending on the order of the two stimuli. KCl produced higher maximal secretion rates and diminished the response to the subsequent glucose stimulus, whereas glucose enhanced the response to the subsequent KCl stimulus. At the level of granule behaviour, a difference developed during the first stimulation phase in that the total number of granules, the short-term resident granules and the arriving granules, which are all parameters of granule turnover, were significantly smaller for glucose than for KCl. These differences at both the level of the cell secretory response and granule behaviour in the submembrane space are incompatible with identical initial response mechanisms to KCl and glucose stimulation.
Collapse
Affiliation(s)
- Dennis Brüning
- Institute of Pharmacology and ToxicologyUniversity of Braunschweig, Braunschweig, Germany
| | - Kirstin Reckers
- Institute of Pharmacology and ToxicologyUniversity of Braunschweig, Braunschweig, Germany
| | - Peter Drain
- Department of Cell BiologyUniversity of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ingo Rustenbeck
- Institute of Pharmacology and ToxicologyUniversity of Braunschweig, Braunschweig, Germany
| |
Collapse
|
26
|
UDP/P2Y6 receptor signaling regulates IgE-dependent degranulation in human basophils. Allergol Int 2017; 66:574-580. [PMID: 28318884 DOI: 10.1016/j.alit.2017.02.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 01/31/2017] [Accepted: 02/13/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND P2Y purinergic receptors (P2YR) are G protein-coupled receptors that are stimulated by extracellular nucleotides. They mediate cellular effects by regulating cAMP production, protein kinase C activation, inositol trisphosphate generation, and Ca2+ release from intracellular stores. The P2Y6 receptor of this family is selectively stimulated by UDP, and selectively inhibited by MRS2578. In the present study, we examined the effect of UDP/P2Y6 receptor signaling on IgE-dependent degranulation in human basophils. METHODS Basophils were purified from human peripheral blood. The mRNA expression of genes encoding P2YR and ecto-nucleoside triphosphate diphosphohydrolase (ENTPDase) was measured by RT-PCR. Intracellular Ca2+ influx via UDP/P2Y6 receptor signaling in basophils was detected using a calcium probe. The effect of UDP/P2Y6 receptor signaling on IgE-dependent degranulation in basophils was confirmed by measuring CD63 expression by flow cytometry. Autocrine secretion of nucleotides was detected by HPLC analysis. RESULTS We showed that purified basophils express P2Y6 mRNA and that UDP increased intracellular Ca2+, which was reduced by MRS2578 treatment. UDP promoted IgE-dependent degranulation. Furthermore, MRS2578 inhibited IgE-dependent degranulation in basophils. HPLC analysis indicated that basophils spontaneously secrete UTP. In addition, basophils expressed the extracellular nucleotide hydrolases ENTPDase2, ENTPDase3, and ENTPDase8. CONCLUSIONS This study showed that UDP/P2Y6 receptor signaling is involved in the regulation of IgE-dependent degranulation in basophils, which might stimulate the P2Y6 receptor via the autocrine secretion of UTP. Thus, this receptor represents a potential target to regulate IgE-dependent degranulation in basophils during allergic diseases.
Collapse
|
27
|
Munc18a clusters SNARE-bearing liposomes prior to trans-SNARE zippering. Biochem J 2017; 474:3339-3354. [PMID: 28827281 DOI: 10.1042/bcj20170494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/16/2017] [Accepted: 08/17/2017] [Indexed: 12/16/2022]
Abstract
Sec1-Munc18 (SM) proteins co-operate with SNAREs {SNAP [soluble NSF (N-ethylmaleimide-sensitive factor) attachment protein] receptors} to mediate membrane fusion in eukaryotic cells. Studies of Munc18a/Munc18-1/Stxbp1 in neurotransmission suggest that SM proteins accelerate fusion kinetics primarily by activating the partially zippered trans-SNARE complex. However, accumulating evidence has argued for additional roles for SM proteins in earlier steps in the fusion cascade. Here, we investigate the function of Munc18a in reconstituted exocytic reactions mediated by neuronal and non-neuronal SNAREs. We show that Munc18a plays a direct role in promoting proteoliposome clustering, underlying vesicle docking during exocytosis. In the three different fusion reactions examined, Munc18a-dependent clustering requires an intact N-terminal peptide (N-peptide) motif in syntaxin that mediates the binary interaction between syntaxin and Munc18a. Importantly, clustering is preserved under inhibitory conditions that abolish both trans-SNARE complex formation and lipid mixing, indicating that Munc18a promotes membrane clustering in a step that is independent of trans-SNARE zippering and activation.
Collapse
|
28
|
Gaisano HY. Recent new insights into the role of SNARE and associated proteins in insulin granule exocytosis. Diabetes Obes Metab 2017; 19 Suppl 1:115-123. [PMID: 28880475 DOI: 10.1111/dom.13001] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 04/23/2017] [Accepted: 05/02/2017] [Indexed: 01/22/2023]
Abstract
Initial work on the exocytotic machinery of predocked insulin secretory granules (SGs) in pancreatic β-cells mimicked the SNARE hypothesis work in neurons, which includes SM/SNARE complex and associated priming proteins, fusion clamps and Ca2+ sensors. However, β-cell SGs, unlike neuronal synaptic vesicles, exhibit a biphasic secretory response that requires additional distinct features in exocytosis including newcomer SGs that undergo minimal docking time at the plasma membrane (PM) before fusion and multi-SG (compound) fusion. These exocytotic events are mediated by Munc18/SNARE complexes distinct from that which mediates predocked SG fusion. We review some recent insights in SNARE complex assembly and the promiscuity in SM/SNARE complex formation, whereby both contribute to conferring different insulin SG fusion kinetics. Some SNARE and associated proteins play non-fusion roles, including tethering SGs to Ca2+ channels, SG recruitment from cell interior to PM, and inhibitory SNAREs that block the action of profusion SNAREs. We discuss new insights into how sub-PM cytoskeletal mesh gates SG access to the PM and the targeting of SG exocytosis to PM domains in functionally polarized β-cells within intact islets. These recent developments have major implications on devising clever SNARE replacement therapies that could restore the deficient insulin secretion in diabetic islet β-cells.
Collapse
|
29
|
Wheeler SE, Stacey HM, Nahaei Y, Hale SJ, Hardy AB, Reimann F, Gribble FM, Larraufie P, Gaisano HY, Brubaker PL. The SNARE Protein Syntaxin-1a Plays an Essential Role in Biphasic Exocytosis of the Incretin Hormone Glucagon-Like Peptide 1. Diabetes 2017; 66:2327-2338. [PMID: 28596237 PMCID: PMC6237272 DOI: 10.2337/db16-1403] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 06/01/2017] [Indexed: 02/02/2023]
Abstract
Exocytosis of the hormone glucagon-like peptide 1 (GLP-1) by the intestinal L cell is essential for the incretin effect after nutrient ingestion and is critical for the actions of dipeptidyl peptidase 4 inhibitors that enhance GLP-1 levels in patients with type 2 diabetes. Two-photon microscopy revealed that exocytosis of GLP-1 is biphasic, with a first peak at 1-6 min and a second peak at 7-12 min after stimulation with forskolin. Approximately 75% of the exocytotic events were represented by compound granule fusion, and the remainder were accounted for by full fusion of single granules under basal and stimulated conditions. The core SNARE protein syntaxin-1a (syn1a) was expressed by murine ileal L cells. At the single L-cell level, first-phase forskolin-induced exocytosis was reduced to basal (P < 0.05) and second-phase exocytosis abolished (P < 0.05) by syn1a knockout. L cells from intestinal-epithelial syn1a-deficient mice demonstrated a 63% reduction in forskolin-induced GLP-1 release in vitro (P < 0.001) and a 23% reduction in oral glucose-stimulated GLP-1 secretion (P < 0.05) in association with impairments in glucose-stimulated insulin release (by 60%; P < 0.01) and glucose tolerance (by 20%; P < 0.01). The findings identify an exquisite mechanism of metered secretory output that precisely regulates release of the incretin hormone GLP-1 and hence insulin secretion after a meal.
Collapse
Affiliation(s)
- Sarah E Wheeler
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Holly M Stacey
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Yasaman Nahaei
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Stephen J Hale
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | | | - Frank Reimann
- Wellcome Trust-MRC Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Cambridge, U.K
| | - Fiona M Gribble
- Wellcome Trust-MRC Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Cambridge, U.K
| | - Pierre Larraufie
- Wellcome Trust-MRC Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Cambridge, U.K
| | - Herbert Y Gaisano
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Patricia L Brubaker
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Duvoor C, Dendi VS, Marco A, Shekhawat NS, Chada A, Ravilla R, Musham CK, Mirza W, Chaudhury A. Commentary: ATP: The crucial component of secretory vesicles: Accelerated ATP/insulin exocytosis and prediabetes. Front Physiol 2017; 8:53. [PMID: 28210227 PMCID: PMC5288386 DOI: 10.3389/fphys.2017.00053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 01/19/2017] [Indexed: 02/03/2023] Open
Affiliation(s)
- Chitharanjan Duvoor
- Department of Endocrinology and Internal Medicine, University of Arkansas for Medical SciencesLittle Rock, AR, USA; GIM FoundationLittle Rock, AR, USA
| | - Vijaya S Dendi
- GIM FoundationLittle Rock, AR, USA; Department of Internal Medicine and Hospital Medicine, Christus Trinity Mother Frances HospitalTyler, TX, USA
| | - Asween Marco
- GIM FoundationLittle Rock, AR, USA; Department of Policy, University of Arkansas for Little RockLittle Rock, AR, USA
| | - Nawal S Shekhawat
- GIM FoundationLittle Rock, AR, USA; Tutwiler ClinicTutwiler, MS, USA
| | - Aditya Chada
- GIM FoundationLittle Rock, AR, USA; Department of Pulmonary and Critical Care Medicine, University of Arkansas for Medical SciencesLittle Rock, AR, USA
| | - Rahul Ravilla
- GIM FoundationLittle Rock, AR, USA; Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical SciencesLittle Rock, AR, USA
| | - Chaitanya K Musham
- GIM FoundationLittle Rock, AR, USA; St. Vincent Infirmary (Catholic Health Initiative)Little Rock, AR, USA
| | | | | |
Collapse
|
31
|
Qin T, Liang T, Zhu D, Kang Y, Xie L, Dolai S, Sugita S, Takahashi N, Ostenson CG, Banks K, Gaisano HY. Munc18b Increases Insulin Granule Fusion, Restoring Deficient Insulin Secretion in Type-2 Diabetes Human and Goto-Kakizaki Rat Islets with Improvement in Glucose Homeostasis. EBioMedicine 2017; 16:262-274. [PMID: 28163042 PMCID: PMC5474508 DOI: 10.1016/j.ebiom.2017.01.030] [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] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 01/09/2017] [Accepted: 01/20/2017] [Indexed: 01/22/2023] Open
Abstract
Reduced pancreatic islet levels of Munc18a/SNARE complex proteins have been postulated to contribute to the deficient glucose-stimulated insulin secretion (GSIS) in type-2 diabetes (T2D). Whereas much previous work has purported Munc18a/SNARE complex (Syntaxin-1A/VAMP-2/SNAP25) to be primarily involved in predocked secretory granule (SG) fusion, less is known about newcomer SGs that undergo minimal docking time at the plasma membrane before fusion. Newcomer SG fusion has been postulated to involve a distinct SM/SNARE complex (Munc18b/Syntaxin-3/VAMP8/SNAP25), whose levels we find also reduced in islets of T2D humans and T2D Goto-Kakizaki (GK) rats. Munc18b overexpression by adenovirus infection (Ad-Munc18b), by increasing assembly of Munc18b/SNARE complexes, mediated increased fusion of not only newcomer SGs but also predocked SGs in T2D human and GK rat islets, resulting in rescue of the deficient biphasic GSIS. Infusion of Ad-Munc18b into GK rat pancreas led to sustained improvement in glucose homeostasis. However, Munc18b overexpression in normal islets increased only newcomer SG fusion. Therefore, Munc18b could potentially be deployed in human T2D to rescue the deficient GSIS. Human T2D islet β-cells exhibit reduced fusion of predocked & newcomer secretory granules (SGs). Munc18b increases SNARE complexes involved in fusions of both newcomer & predocked SGs. Munc18b rescue of newcomer & predocked SGs increased biphasic secretion in human T2D β-cells. Munc18b rescue of T2D Goto-Kakizaki rat β-cell secretion improves glucose homeostasis.
Deficient insulin secretion from pancreatic islet β-cells in type-2 diabetes (T2D) is partly due to reduced expression of many proteins that assemble into specific complexes that mediate fusion of insulin secretory granules (SGs) with plasma membrane, termed exocytosis. We here show we can infuse a virus that contains the construct of one of the SG fusion proteins, Munc18b, into pancreatic ducts of T2D rats to reach the islets, which restored insulin secretion and improved glycemic control. Munc18b acts to promote the assembly of SG fusion complexes. This strategy could potentially be applied to treat human T2D by endoscopic infusion.
Collapse
Affiliation(s)
- Tairan Qin
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Tao Liang
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Dan Zhu
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Youhou Kang
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Li Xie
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Subhankar Dolai
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Shuzo Sugita
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Division of Fundamental Neurobiology, University Health Network, Toronto, ON M5T 2S8, Canada
| | - Noriko Takahashi
- Laboratory of Physiological Chemistry, Institute of Medicinal Chemistry, Hoshi University, Shinagawa, Tokyo 142-8501, Japan
| | - Claes-Goran Ostenson
- Department of Molecular Medicine and Surgery, Endocrinology and Diabetology Unit, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Kate Banks
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Division of Comparative Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Herbert Y Gaisano
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
32
|
Liang T, Qin T, Xie L, Dolai S, Zhu D, Prentice KJ, Wheeler M, Kang Y, Osborne L, Gaisano HY. New Roles of Syntaxin-1A in Insulin Granule Exocytosis and Replenishment. J Biol Chem 2016; 292:2203-2216. [PMID: 28031464 DOI: 10.1074/jbc.m116.769885] [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: 11/26/2016] [Indexed: 01/14/2023] Open
Abstract
In type-2 diabetes (T2D), severely reduced islet syntaxin-1A (Syn-1A) levels contribute to insulin secretory deficiency. We generated β-cell-specific Syn-1A-KO (Syn-1A-βKO) mice to mimic β-cell Syn-1A deficiency in T2D. Glucose tolerance tests showed that Syn-1A-βKO mice exhibited blood glucose elevation corresponding to reduced blood insulin levels. Perifusion of Syn-1A-βKO islets showed impaired first- and second-phase glucose-stimulated insulin secretion (GSIS) resulting from reduction in readily releasable pool and granule pool refilling. To unequivocally determine the β-cell exocytotic defects caused by Syn-1A deletion, EM and total internal reflection fluorescence microscopy showed that Syn-1A-KO β-cells had a severe reduction in the number of secretory granules (SGs) docked onto the plasma membrane (PM) at rest and reduced SG recruitment to the PM after glucose stimulation, the latter indicating defects in replenishment of releasable pools required to sustain second-phase GSIS. Whereas reduced predocked SG fusion accounted for reduced first-phase GSIS, selective reduction of exocytosis of short-dock (but not no-dock) newcomer SGs accounted for the reduced second-phase GSIS. These Syn-1A actions on newcomer SGs were partly mediated by Syn-1A interactions with newcomer SG VAMP8.
Collapse
Affiliation(s)
- Tao Liang
- From the Departments of Medicine.,Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Tairan Qin
- From the Departments of Medicine.,Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Li Xie
- From the Departments of Medicine.,Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Subhankar Dolai
- From the Departments of Medicine.,Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Dan Zhu
- From the Departments of Medicine.,Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Kacey J Prentice
- Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Michael Wheeler
- Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Youhou Kang
- From the Departments of Medicine.,Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Lucy Osborne
- From the Departments of Medicine.,Molecular Genetics, and
| | - Herbert Y Gaisano
- From the Departments of Medicine, .,Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
33
|
Dolai S, Xie L, Zhu D, Liang T, Qin T, Xie H, Kang Y, Chapman ER, Gaisano HY. Synaptotagmin-7 Functions to Replenish Insulin Granules for Exocytosis in Human Islet β-Cells. Diabetes 2016; 65:1962-76. [PMID: 27207520 PMCID: PMC5384637 DOI: 10.2337/db15-1436] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 04/16/2016] [Indexed: 12/13/2022]
Abstract
Synaptotagmin (Syt)-7, a major component of the exocytotic machinery in neurons, is also the major Syt in rodent pancreatic β-cells shown to mediate glucose-stimulated insulin secretion (GSIS). However, Syt-7's precise exocytotic actions in β-cells remain unknown. We show that Syt-7 is abundant in human β-cells. Adenovirus-short hairpin RNA knockdown (KD) of Syt-7 in human islets reduced first- and second-phase GSIS attributed to the reduction of exocytosis of predocked and newcomer insulin secretory granules (SGs). Glucose stimulation expectedly induced Syt-7 association in a Ca(2+)-dependent manner with syntaxin-3 and syntaxin-1A soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes known to mediate exocytosis of newcomer and predocked SGs, respectively. However, Syt-7-KD did not disrupt SNARE complex assembly. Instead, electron microscopy analysis showed that Syt-7-KD reduced the recruitment of SGs to the plasma membrane after glucose-stimulated depletion, which could not be rescued by glucagon-like peptide 1 pretreatment. To assess the possibility that this new action of Syt-7 on SG recruitment may involve calmodulin (CaM), pretreatment of islets with CaM blocker calmidazolium showed effects very similar to those of Syt-7-KD. Syt-7 therefore plays a novel more dominant function in the replenishment of releasable SG pools in human β-cells than its previously purported role in exocytotic fusion per se.
Collapse
Affiliation(s)
- Subhankar Dolai
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Li Xie
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Dan Zhu
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Tao Liang
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Tairan Qin
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Huanli Xie
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Youhou Kang
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Edwin R Chapman
- Department of Neuroscience, Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI
| | - Herbert Y Gaisano
- Department of Medicine, University of Toronto, Toronto, ON, Canada Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
34
|
Hajmrle C, Smith N, Spigelman AF, Dai X, Senior L, Bautista A, Ferdaoussi M, MacDonald PE. Interleukin-1 signaling contributes to acute islet compensation. JCI Insight 2016; 1:e86055. [PMID: 27699257 DOI: 10.1172/jci.insight.86055] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
IL-1β is a well-established inducer of both insulin resistance and impaired pancreatic islet function. Despite this, findings examining IL-1 receptor deficiency or antagonism in in vivo animal models, as well as in clinical studies of type 2 diabetic (T2D) patients, have led to conflicting results, suggesting that the actions of IL-1β on glycemic control may be pleiotropic in nature. In the present work, we find that the ability of IL-1β to amplify glucose-stimulated insulin secretion from human islets correlates with donor BMI. Islets from obese donors are sensitized to the insulinotropic effects of this cytokine, whereas the stimulatory effects of IL-1β are lost in islets from obese T2D patients, suggesting a role for IL-1 signaling in islet compensation. Indeed, mice deficient in IL-1 receptor type I become glucose intolerant more rapidly than their WT littermates and have impaired secretory responses during the acute stages of inflammatory and metabolic stress induced by LPS and high-fat diet, respectively. IL-1β directly enhances β cell insulin secretion by increasing granule docking and soluble N-ethylmaleimide-sensitive factor attachment receptor (SNARE) complex formation at the plasma membrane. Together, our study highlights the importance of IL-1β signaling in islet compensation to metabolic and inflammatory stress.
Collapse
|
35
|
Xie L, Dolai S, Kang Y, Liang T, Xie H, Qin T, Yang L, Chen L, Gaisano HY. Syntaxin-3 Binds and Regulates Both R- and L-Type Calcium Channels in Insulin-Secreting INS-1 832/13 Cells. PLoS One 2016; 11:e0147862. [PMID: 26848587 PMCID: PMC4743851 DOI: 10.1371/journal.pone.0147862] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/08/2016] [Indexed: 12/23/2022] Open
Abstract
Syntaxin (Syn)-1A mediates exocytosis of predocked insulin-containing secretory granules (SGs) during first-phase glucose-stimulated insulin secretion (GSIS) in part via its interaction with plasma membrane (PM)-bound L-type voltage-gated calcium channels (Cav). In contrast, Syn-3 mediates exocytosis of newcomer SGs that accounts for second-phase GSIS. We now hypothesize that the newcomer SG Syn-3 preferentially binds and modulates R-type Cav opening, which was postulated to mediate second-phase GSIS. Indeed, glucose-stimulation of pancreatic islet β-cell line INS-1 induced a predominant increase in interaction between Syn-3 and Cavα1 pore-forming subunits of R-type Cav2.3 and to lesser extent L-type Cavs, while confirming the preferential interactions between Syn-1A with L-type (Cav1.2, Cav1.3) Cavs. Consistently, direct binding studies employing heterologous HEK cells confirmed that Syn-3 preferentially binds Cav2.3, whereas Syn-1A prefers L-type Cavs. We then used siRNA knockdown (KD) of Syn-3 in INS-1 to study the endogenous modulatory actions of Syn-3 on Cav channels. Syn-3 KD enhanced Ca2+ currents by 46% attributed mostly to R- and L-type Cavs. Interestingly, while the transmembrane domain of Syn-1A is the putative functional domain modulating Cav activity, it is the cytoplasmic domain of Syn-3 that appears to modulate Cav activity. We conclude that Syn-3 may mimic Syn-1A in the ability to bind and modulate Cavs, but preferring Cav2.3 to perhaps participate in triggering fusion of newcomer insulin SGs during second-phase GSIS.
Collapse
Affiliation(s)
- Li Xie
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Subhankar Dolai
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Youhou Kang
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Tao Liang
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Huanli Xie
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Tairan Qin
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Lu Yang
- Institute of Molecular Medicine, Peking University, Beijing, China
| | - Liangyi Chen
- Institute of Molecular Medicine, Peking University, Beijing, China
| | - Herbert Y. Gaisano
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- * E-mail:
| |
Collapse
|
36
|
Almaça J, Liang T, Gaisano HY, Nam HG, Berggren PO, Caicedo A. Spatial and temporal coordination of insulin granule exocytosis in intact human pancreatic islets. Diabetologia 2015; 58:2810-8. [PMID: 26376795 PMCID: PMC6132229 DOI: 10.1007/s00125-015-3747-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/12/2015] [Indexed: 01/13/2023]
Abstract
AIMS/HYPOTHESIS Insulin secretion is widely studied because it plays a central role in glucose homeostasis and diabetes. Processes from insulin granule fusion in beta cells to in vivo insulin secretion have been elucidated, but data at the cellular level do not fully account for several aspects of the macroscopic secretory pattern. Here we investigated how individual secretory events are coordinated spatially and temporally within intact human islets. METHODS We used the fluorescent probe neuropeptide Y (NPY)-pHluorin to visualise insulin granule secretion in isolated intact human islets. RESULTS We found that individual beta cells respond to increases in glucose concentration by releasing insulin granules in very discrete bursts with periods consistent with in vivo pulsatile insulin secretion. In successive secretory bursts during prolonged exposure to high glucose levels, secretory events progressively localised to preferential release sites, coinciding with the transition to second phase insulin secretion. Granule secretion was very synchronised in neighbouring beta cells, forming discrete regional clusters of activity. CONCLUSIONS/INTERPRETATION These results reveal how individual secretory events are coordinated to produce pulsatile insulin secretion from human islets.
Collapse
Affiliation(s)
- Joana Almaça
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL, 33136, USA.
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA.
- Center for Plant Aging Research, Institute for Basic Science and Department of New Biology, DGIST, Daegu, Republic of Korea.
| | - Tao Liang
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Hong Gil Nam
- Center for Plant Aging Research, Institute for Basic Science and Department of New Biology, DGIST, Daegu, Republic of Korea
| | - Per-Olof Berggren
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL, 33136, USA.
- Rolf Luft Research Center for Diabetes & Endocrinology, Karolinska Institutet, Stockholm, Sweden.
- Lee Kong Chien School of Medicine, Nanyang Technical University, Singapore, Singapore.
- Imperial College, London, UK.
| | - Alejandro Caicedo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL, 33136, USA.
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, FL, USA.
- Program in Neuroscience, Miller School of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
37
|
β-Cell Insulin Secretion Requires the Ubiquitin Ligase COP1. Cell 2015; 163:1457-67. [PMID: 26627735 DOI: 10.1016/j.cell.2015.10.076] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 09/22/2015] [Accepted: 10/19/2015] [Indexed: 11/20/2022]
Abstract
A variety of signals finely tune insulin secretion by pancreatic β cells to prevent both hyper-and hypoglycemic states. Here, we show that post-translational regulation of the transcription factors ETV1, ETV4, and ETV5 by the ubiquitin ligase COP1 (also called RFWD2) in β cells is critical for insulin secretion. Mice lacking COP1 in β cells developed diabetes due to insulin granule docking defects that were fully rescued by genetic deletion of Etv1, Etv4, and Etv5. Genes regulated by ETV1, ETV4, or ETV5 in the absence of mouse COP1 were enriched in human diabetes-associated genes, suggesting that they also influence human β-cell pathophysiology. In normal β cells, ETV4 was stabilized upon membrane depolarization and limited insulin secretion under hyperglycemic conditions. Collectively, our data reveal that ETVs negatively regulate insulin secretion for the maintenance of normoglycemia.
Collapse
|
38
|
Xu H, Abuhatzira L, Carmona GN, Vadrevu S, Satin LS, Notkins AL. The Ia-2β intronic miRNA, miR-153, is a negative regulator of insulin and dopamine secretion through its effect on the Cacna1c gene in mice. Diabetologia 2015; 58:2298-306. [PMID: 26141787 PMCID: PMC6754265 DOI: 10.1007/s00125-015-3683-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 06/11/2015] [Indexed: 12/23/2022]
Abstract
AIMS/HYPOTHESIS miR-153 is an intronic miRNA embedded in the genes that encode IA-2 (also known as PTPRN) and IA-2β (also known as PTPRN2). Islet antigen (IA)-2 and IA-2β are major autoantigens in type 1 diabetes and are important transmembrane proteins in dense core and synaptic vesicles. miR-153 and its host genes are co-regulated in pancreas and brain. The present experiments were initiated to decipher the regulatory network between miR-153 and its host gene Ia-2β (also known as Ptprn2). METHODS Insulin secretion was determined by ELISA. Identification of miRNA targets was assessed using luciferase assays and by quantitative real-time PCR and western blots in vitro and in vivo. Target protector was also employed to evaluate miRNA target function. RESULTS Functional studies revealed that miR-153 mimic suppresses both glucose- and potassium-induced insulin secretion (GSIS and PSIS, respectively), whereas miR-153 inhibitor enhances both GSIS and PSIS. A similar effect on dopamine secretion also was observed. Using miRNA target prediction software, we found that miR-153 is predicted to target the 3'UTR region of the calcium channel gene, Cacna1c. Further studies confirmed that Cacna1c mRNA and protein are downregulated by miR-153 mimics and upregulated by miR-153 inhibitors in insulin-secreting freshly isolated mouse islets, in the insulin-secreting mouse cell line MIN6 and in the dopamine-secreting cell line PC12. CONCLUSIONS/INTERPRETATION miR-153 is a negative regulator of both insulin and dopamine secretion through its effect on Cacna1c expression, which suggests that IA-2β and miR-153 have opposite functional effects on the secretory pathway.
Collapse
Affiliation(s)
- Huanyu Xu
- Experimental Medicine Section, Laboratory of Sensory Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Liron Abuhatzira
- Experimental Medicine Section, Laboratory of Sensory Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Gilberto N Carmona
- Experimental Medicine Section, Laboratory of Sensory Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Suryakiran Vadrevu
- Brehm Diabetes Research Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Leslie S Satin
- Brehm Diabetes Research Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Abner L Notkins
- Experimental Medicine Section, Laboratory of Sensory Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA.
| |
Collapse
|
39
|
Fan F, Ji C, Wu Y, Ferguson SM, Tamarina N, Philipson LH, Lou X. Dynamin 2 regulates biphasic insulin secretion and plasma glucose homeostasis. J Clin Invest 2015; 125:4026-41. [PMID: 26413867 DOI: 10.1172/jci80652] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 08/20/2015] [Indexed: 12/18/2022] Open
Abstract
Alterations in insulin granule exocytosis and endocytosis are paramount to pancreatic β cell dysfunction in diabetes mellitus. Here, using temporally controlled gene ablation specifically in β cells in mice, we identified an essential role of dynamin 2 GTPase in preserving normal biphasic insulin secretion and blood glucose homeostasis. Dynamin 2 deletion in β cells caused glucose intolerance and substantial reduction of the second phase of glucose-stimulated insulin secretion (GSIS); however, mutant β cells still maintained abundant insulin granules, with no signs of cell surface expansion. Compared with control β cells, real-time capacitance measurements demonstrated that exocytosis-endocytosis coupling was less efficient but not abolished; clathrin-mediated endocytosis (CME) was severely impaired at the step of membrane fission, which resulted in accumulation of clathrin-coated endocytic intermediates on the plasma membrane. Moreover, dynamin 2 ablation in β cells led to striking reorganization and enhancement of actin filaments, and insulin granule recruitment and mobilization were impaired at the later stage of GSIS. Together, our results demonstrate that dynamin 2 regulates insulin secretory capacity and dynamics in vivo through a mechanism depending on CME and F-actin remodeling. Moreover, this study indicates a potential pathophysiological link between endocytosis and diabetes mellitus.
Collapse
|
40
|
Schwede F, Chepurny OG, Kaufholz M, Bertinetti D, Leech CA, Cabrera O, Zhu Y, Mei F, Cheng X, Manning Fox JE, MacDonald PE, Genieser HG, Herberg FW, Holz GG. Rp-cAMPS Prodrugs Reveal the cAMP Dependence of First-Phase Glucose-Stimulated Insulin Secretion. Mol Endocrinol 2015; 29:988-1005. [PMID: 26061564 DOI: 10.1210/me.2014-1330] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
cAMP-elevating agents such as the incretin hormone glucagon-like peptide-1 potentiate glucose-stimulated insulin secretion (GSIS) from pancreatic β-cells. However, a debate has existed since the 1970s concerning whether or not cAMP signaling is essential for glucose alone to stimulate insulin secretion. Here, we report that the first-phase kinetic component of GSIS is cAMP-dependent, as revealed through the use of a novel highly membrane permeable para-acetoxybenzyl (pAB) ester prodrug that is a bioactivatable derivative of the cAMP antagonist adenosine-3',5'-cyclic monophosphorothioate, Rp-isomer (Rp-cAMPS). In dynamic perifusion assays of human or rat islets, a step-wise increase of glucose concentration leads to biphasic insulin secretion, and under these conditions, 8-bromoadenosine-3',5'-cyclic monophosphorothioate, Rp-isomer, 4-acetoxybenzyl ester (Rp-8-Br-cAMPS-pAB) inhibits first-phase GSIS by up to 80%. Surprisingly, second-phase GSIS is inhibited to a much smaller extent (≤20%). Using luciferase, fluorescence resonance energy transfer, and bioluminescence resonance energy transfer assays performed in living cells, we validate that Rp-8-Br-cAMPS-pAB does in fact block cAMP-dependent protein kinase activation. Novel effects of Rp-8-Br-cAMPS-pAB to block the activation of cAMP-regulated guanine nucleotide exchange factors (Epac1, Epac2) are also validated using genetically encoded Epac biosensors, and are independently confirmed in an in vitro Rap1 activation assay using Rp-cAMPS and Rp-8-Br-cAMPS. Thus, in addition to revealing the cAMP dependence of first-phase GSIS from human and rat islets, these findings establish a pAB-based chemistry for the synthesis of highly membrane permeable prodrug derivatives of Rp-cAMPS that act with micromolar or even nanomolar potency to inhibit cAMP signaling in living cells.
Collapse
Affiliation(s)
- Frank Schwede
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Oleg G Chepurny
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Melanie Kaufholz
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Daniela Bertinetti
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Colin A Leech
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Over Cabrera
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Yingmin Zhu
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Fang Mei
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Xiaodong Cheng
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Jocelyn E Manning Fox
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Patrick E MacDonald
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Hans-G Genieser
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - Friedrich W Herberg
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| | - George G Holz
- BIOLOG Life Science Institute (F.S., H.-G.G.), 28199 Bremen, Germany; Departments of Medicine (O.G.C., C.A.L., G.G.H.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Department of Biochemistry (M.K., D.B., F.W.H.), University of Kassel, 34132 Kassel, Germany; Eli Lilly and Company (O.C.), Indianapolis, Indiana 46225; Department of Integrative Biology and Pharmacology (Y.Z., F.M., X.C.), Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, Texas 77030; Department of Pharmacology and the Alberta Diabetes Institute (J.E.M.F., P.E.M.), University of Alberta, Edmonton, Canada AB T6G 2E1
| |
Collapse
|
41
|
Zhu D, Xie L, Karimian N, Liang T, Kang Y, Huang YC, Gaisano HY. Munc18c mediates exocytosis of pre-docked and newcomer insulin granules underlying biphasic glucose stimulated insulin secretion in human pancreatic beta-cells. Mol Metab 2015; 4:418-26. [PMID: 25973389 PMCID: PMC4421095 DOI: 10.1016/j.molmet.2015.02.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 02/05/2015] [Accepted: 02/09/2015] [Indexed: 01/10/2023] Open
Abstract
Objective Pancreatic beta-cells express three Munc18 isoforms. Much is known about the roles of Munc18a (pre-docked secretory granules-SGs) and Munc18b (newcomer SGs and SG–SG fusion) in insulin exocytosis. Although shown to influence glucose-stimulated insulin secretion (GSIS) in rodents the precise role of Munc18c in insulin SG exocytosis has not been elucidated. We here examined the role of Munc18c in human pancreatic beta-cells. Methods Munc18c-shRNA/RFP lenti-virus (versus control virus) was used to knock down the expression level of Munc18c in human islets or single beta-cells. Insulin secretion and granule exocytosis were measured by performing islets perifusion, single-cell patch clamp capacitance measurements and total internal reflection fluorescence microscopy (TIRFM). Results Munc18c is most abundant in the cytosol of human beta-cells. Endogenous function of Munc18c was assessed by knocking down (KD) its islet expression by 70% employing lenti-shRNA virus. Munc18c-KD caused reduction in cognate syntaxin-4 islet expression but not in other exocytotic proteins, resulting in the reduction in GSIS in first- (by 42%) and second phases (by 35%). Single cell analyses of RFP-tagged Munc18c-KD beta-cells by patch clamp capacitance measurements showed inhibition in both readily-releasable pool (by 52%) and mobilization from the reserve pool (by 57%). TIRFM to assess single SG behavior showed that Munc18c-KD inhibition of first phase GSIS was attributed to reduction in exocytosis of pre-docked and newcomer SGs, and second phase inhibition attributed entirely to reduction in newcomer SG fusion (SGs that undergo minimal residence or docking time at the plasma membrane before fusion). Conclusion Munc18c is involved in the distinct molecular machineries that affect exocytosis of both predocked and newcomer SG pools that underlie Munc18c's role in first and second phases of GSIS, respectively.
Collapse
Key Words
- Ad, adenovirus
- CmPatch, clamp capacitance measurements
- EGFP, enhanced green fluorescent protein
- Exocytosis
- GLP-1, glucagon-like peptide-1
- GSIS, glucose-stimulated insulin secretion
- Human islets
- KD, knock down
- Munc18c
- NPY, neuropeptide Y
- Newcomer insulin granules
- PM, plasma membrane
- RRP, readily releasable pool
- SG, secretory insulin-containing granule
- SM, Sec1/Munc18-like protein
- SNAP25/23, synaptosomal-associated protein of 25/23 kD
- SNARE, soluble N-ethylmaleimide-sensitive factor attachment protein receptor
- Syn, syntaxin
- T2DM, type 2 diabetes mellitus
- TIRFM, total internal reflection fluorescence microscopy
- VAMPs, Vesicle Associated Membrane Proteins
- t-, target-
- v-, vesicle-
Collapse
Affiliation(s)
- Dan Zhu
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Li Xie
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Negar Karimian
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Tao Liang
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Youhou Kang
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Ya-Chi Huang
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Herbert Y Gaisano
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
42
|
Li H, Liu T, Lim J, Gounko NV, Hong W, Han W. Increased biogenesis of glucagon-containing secretory granules and glucagon secretion in BIG3-knockout mice. Mol Metab 2015; 4:246-52. [PMID: 25737957 PMCID: PMC4338310 DOI: 10.1016/j.molmet.2015.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 12/24/2014] [Accepted: 01/03/2015] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE Although both insulin and glucagon are intimately involved in the regulation of glucose homeostasis, the intrinsic control of glucagon secretion, including the biogenesis and exocytosis of glucagon-containing granules, is far less understood compared with that of insulin. As Brefeldin A-inhibited guanine nucleotide exchange protein 3 (BIG3) is a negative regulator of insulin-granule biogenesis and insulin secretion, we investigated whether BIG3 plays any role in alpha-cells and glucagon secretion. METHODS We examined the expression of BIG3 in islet cells by immuno-fluorescence and confocal microscopy, and measured glucagon production and secretion in BIG3-depleted and wild-type mice, islets and cells. RESULTS BIG3 is highly expressed in pancreatic alpha-cells in addition to beta-cells, but is absent in delta-cells. Depletion of BIG3 in alpha-cells leads to elevated glucagon production and secretion. Consistently, BIG3-knockout (BKO) mice display increased glucagon release under hypoglycemic conditions. CONCLUSIONS Together with our previous studies, the current data reveal a conserved role for BIG3 in regulating alpha- and beta-cell functions. We propose that BIG3 negatively regulates hormone production at the secretory granule biogenesis stage and that such regulatory mechanism may be used in secretory pathways of other endocrine cells.
Collapse
Affiliation(s)
- Hongyu Li
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (ASTAR), #02-02 Helios, 11 Biopolis Way, Singapore 138667, Singapore
| | - Tao Liu
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (ASTAR), #02-02 Helios, 11 Biopolis Way, Singapore 138667, Singapore
| | - Joy Lim
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (ASTAR), #02-02 Helios, 11 Biopolis Way, Singapore 138667, Singapore
| | - Natalia V Gounko
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore 138667, Singapore ; Joint IMB-IMCB Electron Microscopy Suite, Agency for Science, Technology and Research (A*STAR), Singapore 138667, Singapore
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore 138667, Singapore
| | - Weiping Han
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (ASTAR), #02-02 Helios, 11 Biopolis Way, Singapore 138667, Singapore
| |
Collapse
|
43
|
Liu T, Li H, Gounko NV, Zhou Z, Xu A, Hong W, Han W. Detection of insulin granule exocytosis by an electrophysiology method with high temporal resolution reveals enlarged insulin granule pool in BIG3-knockout mice. Am J Physiol Endocrinol Metab 2014; 307:E611-8. [PMID: 25139048 DOI: 10.1152/ajpendo.00208.2014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We recently identified BIG3 as a negative regulator of insulin granule biogenesis and reported increased insulin secretion in BIG3-knockout (BKO) mice. To pinpoint the site of action for BIG3, we investigated whether BIG3 regulates quantal insulin granule exocytosis. We established an assay to detect insulin granule exocytosis by recording ATP-elicited currents at high temporal resolution by patch clamp. Similarly to insulin, ATP release was increased in BKO β-cells. Although the frequency of insulin granule exocytosis was increased in BKO β-cells, quantal size or release kinetics remained unchanged. Electron microscopy studies showed that the number of insulin granules was increased by >60% in BKO β-cells. However, the number of morphologically docked granules was unaltered. The number of insulin granules having significant distances away from plasma membrane was greatly increased in BKO β-cells. Thus, BIG3 negatively regulates insulin granule exocytosis by restricting insulin granule biogenesis without the release kinetics of individual granules at the final exocytotic steps being affected. Depletion of BIG3 leads to an enlarged releasable pool of insulin granules, which accounts for increased release frequency and consequently increased insulin secretion.
Collapse
Affiliation(s)
- Tao Liu
- Singapore Bioimaging Consortium
| | - Hongyu Li
- Singapore Bioimaging Consortium, Institute of Molecular and Cell Biology, and
| | - Natalia V Gounko
- Institute of Molecular and Cell Biology, and Joint IMB-IMCB Electron Microscopy Suite, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Zhuan Zhou
- Institute of Molecular Medicine, Peking University, Peking, China; and
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, and Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, and
| | | |
Collapse
|
44
|
Henquin JC, Nenquin M. Activators of PKA and Epac distinctly influence insulin secretion and cytosolic Ca2+ in female mouse islets stimulated by glucose and tolbutamide. Endocrinology 2014; 155:3274-87. [PMID: 24977470 PMCID: PMC4255079 DOI: 10.1210/en.2014-1247] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Amplification of insulin secretion by cAMP is mediated by protein kinase A (PKA) and exchange protein directly activated by cAMP (Epac). Using selective activators, we determined how each effector influences the cytosolic free Ca(2+) concentration ([Ca(2+)]c) and insulin secretion in mouse islets. Alone PKA activator amplified glucose- and tolbutamide-induced insulin secretion, with a greater impact on second than first phase. Epac activator strongly amplified both phases in response to either secretagogue. Amplification was even greater when activators were combined. Although both activators similarly amplified glucose-induced insulin secretion, Epac activator was particularly efficient on tolbutamide-induced insulin secretion. That greater efficacy is attributed to higher [Ca(2+)]c rather than interaction of tolbutamide with Epac, because it was also observed during KCl stimulation. Moreover, in contrast to Epac activator, tolbutamide was inactive when insulin secretion was increased by gliclazide, and its effect on glucose-induced insulin secretion was unaffected by an inhibitor of Epac2. PKA activator increased [Ca(2+)]c during acute or steady-state glucose stimulation, whereas Epac activator had no effect alone or in combination. Neither activator affected [Ca(2+)]c response to tolbutamide or KCl. Metabolic (glucose-mediated) amplification of insulin secretion was unaffected by PKA activator. It was attenuated when insulin secretion was augmented by Epac activator but insensitive to Epac2 inhibitor, which suggests distinct although somewhat overlapping mechanisms. In conclusion, activators of PKA and Epac amplify insulin secretion by augmenting the action of Ca(2+) on exocytosis and, for PKA only, slightly increasing glucose-induced [Ca(2+)]c rise. The influence of Epac seems more important than that of PKA during first phase.
Collapse
Affiliation(s)
- Jean-Claude Henquin
- Unit of Endocrinology and Metabolism, Faculty of Medicine, Université Catholique de Louvain, B-1200 Brussels, Belgium
| | | |
Collapse
|