1
|
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
|
2
|
Salazar Lázaro A, Trimbuch T, Vardar G, Rosenmund C. The stability of the primed pool of synaptic vesicles and the clamping of spontaneous neurotransmitter release rely on the integrity of the C-terminal half of the SNARE domain of syntaxin-1A. eLife 2024; 12:RP90775. [PMID: 38512129 PMCID: PMC10957171 DOI: 10.7554/elife.90775] [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] [Indexed: 03/22/2024] Open
Abstract
The SNARE proteins are central in membrane fusion and, at the synapse, neurotransmitter release. However, their involvement in the dual regulation of the synchronous release while maintaining a pool of readily releasable vesicles remains unclear. Using a chimeric approach, we performed a systematic analysis of the SNARE domain of STX1A by exchanging the whole SNARE domain or its N- or C-terminus subdomains with those of STX2. We expressed these chimeric constructs in STX1-null hippocampal mouse neurons. Exchanging the C-terminal half of STX1's SNARE domain with that of STX2 resulted in a reduced RRP accompanied by an increased release rate, while inserting the C-terminal half of STX1's SNARE domain into STX2 leads to an enhanced priming and decreased release rate. Additionally, we found that the mechanisms for clamping spontaneous, but not for Ca2+-evoked release, are particularly susceptible to changes in specific residues on the outer surface of the C-terminus of the SNARE domain of STX1A. Particularly, mutations of D231 and R232 affected the fusogenicity of the vesicles. We propose that the C-terminal half of the SNARE domain of STX1A plays a crucial role in the stabilization of the RRP as well as in the clamping of spontaneous synaptic vesicle fusion through the regulation of the energetic landscape for fusion, while it also plays a covert role in the speed and efficacy of Ca2+-evoked release.
Collapse
Affiliation(s)
- Andrea Salazar Lázaro
- Department of Neurophysiology, Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin Institute of HealthBerlinGermany
| | - Thorsten Trimbuch
- Department of Neurophysiology, Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin Institute of HealthBerlinGermany
| | - Gülçin Vardar
- Department of Neurophysiology, Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin Institute of HealthBerlinGermany
| | - Christian Rosenmund
- Department of Neurophysiology, Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin Institute of HealthBerlinGermany
- NeuroCure Excellence ClusterBerlinGermany
| |
Collapse
|
3
|
Wang H, Li Q, Yuan YC, Han XC, Cao YT, Yang JK. KCNH6 channel promotes insulin exocytosis via interaction with Munc18-1 independent of electrophysiological processes. Cell Mol Life Sci 2024; 81:86. [PMID: 38349432 PMCID: PMC10864572 DOI: 10.1007/s00018-024-05134-1] [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: 09/30/2023] [Revised: 12/23/2023] [Accepted: 01/19/2024] [Indexed: 02/15/2024]
Abstract
Glucose-stimulated insulin secretion (GSIS) in pancreatic islet β-cells primarily relies on electrophysiological processes. Previous research highlighted the regulatory role of KCNH6, a member of the Kv channel family, in governing GSIS through its influence on β-cell electrophysiology. In this study, we unveil a novel facet of KCNH6's function concerning insulin granule exocytosis, independent of its conventional electrical role. Young mice with β-cell-specific KCNH6 knockout (βKO) exhibited impaired glucose tolerance and reduced insulin secretion, a phenomenon not explained by electrophysiological processes alone. Consistently, islets from KCNH6-βKO mice exhibited reduced insulin secretion, conversely, the overexpression of KCNH6 in murine pancreatic islets significantly enhanced insulin release. Moreover, insulin granules lacking KCNH6 demonstrated compromised docking capabilities and a reduced fusion response upon glucose stimulation. Crucially, our investigation unveiled a significant interaction between KCNH6 and the SNARE protein regulator, Munc18-1, a key mediator of insulin granule exocytosis. These findings underscore the critical role of KCNH6 in the regulation of insulin secretion through its interaction with Munc18-1, providing a promising and novel avenue for enhancing our understanding of the Kv channel in diabetes mechanisms.
Collapse
Affiliation(s)
- Hao Wang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology and Metabolism, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China.
| | - Qi Li
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology and Metabolism, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
| | - Ying-Chao Yuan
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology and Metabolism, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Xue-Chun Han
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology and Metabolism, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Yong-Ting Cao
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology and Metabolism, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
- Department of Endocrinology, Beijing Mentougou District Hospital, Beijing, 102399, China
| | - Jin-Kui Yang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology and Metabolism, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
4
|
Moss ND, Wells KL, Theis A, Kim YK, Spigelman AF, Liu X, MacDonald PE, Sussel L. Modulation of insulin secretion by RBFOX2-mediated alternative splicing. Nat Commun 2023; 14:7732. [PMID: 38007492 PMCID: PMC10676425 DOI: 10.1038/s41467-023-43605-4] [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: 01/31/2023] [Accepted: 11/15/2023] [Indexed: 11/27/2023] Open
Abstract
Insulin secretion is a tightly regulated process that is vital for maintaining blood glucose homeostasis. Although the molecular components of insulin granule trafficking and secretion are well established, how they are regulated to rapidly fine-tune secretion in response to changing environmental conditions is not well characterized. Recent studies have determined that dysregulation of RNA-binding proteins (RBPs) and aberrant mRNA splicing occurs at the onset of diabetes. We demonstrate that the RBP, RBFOX2, is a critical regulator of insulin secretion through the alternative splicing of genes required for insulin granule docking and exocytosis. Conditional mutation of Rbfox2 in the mouse pancreas results in decreased insulin secretion and impaired blood glucose homeostasis. Consistent with defects in secretion, we observe reduced insulin granule docking and corresponding splicing defects in the SNARE complex components. These findings identify an additional mechanism for modulating insulin secretion in both healthy and dysfunctional pancreatic β cells.
Collapse
Affiliation(s)
- Nicole D Moss
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kristen L Wells
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alexandra Theis
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Yong-Kyung Kim
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Aliya F Spigelman
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Xiong Liu
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Patrick E MacDonald
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Lori Sussel
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
5
|
Vinci M, Costanza C, Galati Rando R, Treccarichi S, Saccone S, Carotenuto M, Roccella M, Calì F, Elia M, Vetri L. STXBP6 Gene Mutation: A New Form of SNAREopathy Leads to Developmental Epileptic Encephalopathy. Int J Mol Sci 2023; 24:16436. [PMID: 38003627 PMCID: PMC10670990 DOI: 10.3390/ijms242216436] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
Syntaxin-binding protein 6 (STXBP6), also known as amysin, is an essential component of the SNAP receptor (SNARE) complex and plays a crucial role in neuronal vesicle trafficking. Mutations in genes encoding SNARE proteins are often associated with a broad spectrum of neurological conditions defined as "SNAREopathies", including epilepsy, intellectual disability, and neurodevelopmental disorders such as autism spectrum disorders. The present whole exome sequencing (WES) study describes, for the first time, the occurrence of developmental epileptic encephalopathy and autism spectrum disorders as a result of a de novo deletion within the STXBP6 gene. The truncated protein in the STXBP6 gene leading to a premature stop codon could negatively modulate the synaptic vesicles' exocytosis. Our research aimed to elucidate a plausible, robust correlation between STXBP6 gene deletion and the manifestation of developmental epileptic encephalopathy.
Collapse
Affiliation(s)
- Mirella Vinci
- Oasi Research Institute-IRCCS, 94018 Troina, Italy; (M.V.); (R.G.R.); (S.T.); (M.E.); (L.V.)
| | - Carola Costanza
- Department of Psychology, Educational Science and Human Movement, University of Palermo, 90141 Palermo, Italy; (C.C.); (M.R.)
| | - Rosanna Galati Rando
- Oasi Research Institute-IRCCS, 94018 Troina, Italy; (M.V.); (R.G.R.); (S.T.); (M.E.); (L.V.)
| | - Simone Treccarichi
- Oasi Research Institute-IRCCS, 94018 Troina, Italy; (M.V.); (R.G.R.); (S.T.); (M.E.); (L.V.)
| | - Salvatore Saccone
- Department Biological, Geological and Environmental Sciences, University of Catania, Via Androne 81, 95124 Catania, Italy;
| | - Marco Carotenuto
- Clinic of Child and Adolescent Neuropsychiatry, Department of Mental Health, Physical and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy;
| | - Michele Roccella
- Department of Psychology, Educational Science and Human Movement, University of Palermo, 90141 Palermo, Italy; (C.C.); (M.R.)
| | - Francesco Calì
- Oasi Research Institute-IRCCS, 94018 Troina, Italy; (M.V.); (R.G.R.); (S.T.); (M.E.); (L.V.)
| | - Maurizio Elia
- Oasi Research Institute-IRCCS, 94018 Troina, Italy; (M.V.); (R.G.R.); (S.T.); (M.E.); (L.V.)
| | - Luigi Vetri
- Oasi Research Institute-IRCCS, 94018 Troina, Italy; (M.V.); (R.G.R.); (S.T.); (M.E.); (L.V.)
| |
Collapse
|
6
|
Tsuyama T, Sato Y, Yoshizawa T, Matsuoka T, Yamagata K. Hypoxia causes pancreatic β-cell dysfunction and impairs insulin secretion by activating the transcriptional repressor BHLHE40. EMBO Rep 2023; 24:e56227. [PMID: 37341148 PMCID: PMC10398664 DOI: 10.15252/embr.202256227] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 05/28/2023] [Accepted: 06/04/2023] [Indexed: 06/22/2023] Open
Abstract
Hypoxia can occur in pancreatic β-cells in type 2 diabetes. Although hypoxia exerts deleterious effects on β-cell function, the associated mechanisms are largely unknown. Here, we show that the transcriptional repressor basic helix-loop-helix family member e40 (BHLHE40) is highly induced in hypoxic mouse and human β-cells and suppresses insulin secretion. Conversely, BHLHE40 deficiency in hypoxic MIN6 cells or β-cells of ob/ob mice reverses defects in insulin secretion. Mechanistically, BHLHE40 represses the expression of Mafa, encoding the transcription factor musculoaponeurotic fibrosarcoma oncogene family A (MAFA), by attenuating the binding of pancreas/duodenum homeobox protein 1 (PDX1) to its enhancer region. Impaired insulin secretion in hypoxic β-cells was recovered by MAFA re-expression. Collectively, our work identifies BHLHE40 as a key hypoxia-induced transcriptional repressor in β-cells that inhibit insulin secretion by suppressing MAFA expression.
Collapse
Affiliation(s)
- Tomonori Tsuyama
- Center for Metabolic Regulation of Healthy Aging (CMHA), Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Yoshifumi Sato
- Department of Medical Biochemistry, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Tatsuya Yoshizawa
- Department of Medical Biochemistry, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Takaaki Matsuoka
- First Department of Internal MedicineWakayama Medical UniversityWakayamaJapan
| | - Kazuya Yamagata
- Center for Metabolic Regulation of Healthy Aging (CMHA), Faculty of Life SciencesKumamoto UniversityKumamotoJapan
- Department of Medical Biochemistry, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| |
Collapse
|
7
|
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
|
8
|
Thoduvayil S, Weerakkody JS, Sundaram RVK, Topper M, Bera M, Coleman J, Li X, Mariappan M, Ramakrishnan S. Rapid Quantification of First and Second Phase Insulin Secretion Dynamics using an In vitro Platform for Improving Insulin Therapy. Cell Calcium 2023; 113:102766. [PMID: 37295201 PMCID: PMC10450995 DOI: 10.1016/j.ceca.2023.102766] [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: 04/03/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023]
Abstract
High-throughput quantification of the first- and second-phase insulin secretion dynamics is intractable with current methods. The fact that independent secretion phases play distinct roles in metabolism necessitates partitioning them separately and performing high-throughput compound screening to target them individually. We developed an insulin-nanoluc luciferase reporter system to dissect the molecular and cellular pathways involved in the separate phases of insulin secretion. We validated this method through genetic studies, including knockdown and overexpression, as well as small-molecule screening and their effects on insulin secretion. Furthermore, we demonstrated that the results of this method are well correlated with those of single-vesicle exocytosis experiments conducted on live cells, providing a quantitative reference for the approach. Thus, we have developed a robust methodology for screening small molecules and cellular pathways that target specific phases of insulin secretion, resulting in a better understanding of insulin secretion, which in turn will result in a more effective insulin therapy through the stimulation of endogenous glucose-stimulated insulin secretion.
Collapse
Affiliation(s)
- Sikha Thoduvayil
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Pathology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Jonathan S Weerakkody
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Pathology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Ramalingam Venkat Kalyana Sundaram
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Mackenzie Topper
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA
| | - Manindra Bera
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Jeff Coleman
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Xia Li
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Malaiyalam Mariappan
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Sathish Ramakrishnan
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Pathology, Yale University School of Medicine, New Haven, CT, 06520 USA.
| |
Collapse
|
9
|
Li W, Xing Y, Wang Y, Xu T, Song E, Feng W. A non-canonical target-binding site in Munc18-1 domain 3b for assembling the Mint1-Munc18-1-syntaxin-1 complex. Structure 2023; 31:68-77.e5. [PMID: 36608665 DOI: 10.1016/j.str.2022.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/10/2022] [Accepted: 11/04/2022] [Indexed: 01/07/2023]
Abstract
As the prototype of Sec1/Munc18 (SM) family proteins, Munc18-1 can manipulate the distinct conformations of syntaxin-1 for controlling intracellular membrane fusion. The Munc18-1-interacting domain of Mint1 (Mint1-MID) binds to Munc18-1 together with syntaxin-1 to form a Mint1-Munc18-1-syntaxin-1 complex, but the mechanism underlying the complex assembly remains unclear. Here, we determine the structure of the Mint1-MID-Munc18-1-syntaxin-1 complex. Unexpectedly, Munc18-1 recognizes Mint1-MID and syntaxin-1 simultaneously via two opposite sites. The canonical central cavity between domains 1 and 3a of Munc18-1 embraces closed syntaxin-1, whereas the non-canonical basic pocket in domain 3b captures the acidic Mint1-MID helix. The domain 3b-mediated recognition of an acidic-helical motif is distinct from other target-recognition modes of Munc18-1. Mutations in the interface between domain 3b and Mint1-MID disrupt the assembly of the Mint1-Munc18-1-syntaxin-1 complex. This work reveals a non-canonical target-binding site in Munc18-1 domain 3b for assembling the Mint1-Munc18-1-syntaxin-1 complex.
Collapse
Affiliation(s)
- Wei Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China
| | - Ying Xing
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yue Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tao Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Eli Song
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China.
| | - Wei Feng
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
10
|
Zhu M, Xu H, Jiang Y, Yu H, Liu Y. Epigallocatechin gallate inhibits SNARE-dependent membrane fusion by blocking trans-SNARE assembly. FEBS Open Bio 2022; 12:2111-2121. [PMID: 36111501 PMCID: PMC9714361 DOI: 10.1002/2211-5463.13488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/06/2022] [Accepted: 09/15/2022] [Indexed: 01/25/2023] Open
Abstract
Insulin secretion is a signal-triggered process that requires membrane fusion between the secretory granules and plasma membrane in pancreatic β cells. The exocytosis of insulin is mediated by target-soluble N-ethylmaleimide sensitive factor attachment protein receptors (SNAREs) on the plasma membrane and vesicle-SNAREs on the vesicles, which assemble into a quaternary trans-SNARE complex to initiate the fusion. Expression of fusion proteins is reduced in the islets of patients with type II diabetes, indicating that SNARE-mediated fusion defect is closely related to insulin-based metabolic diseases. Previous studies have suggested that epigallocatechin gallate (EGCG) has an inhibitory effect on membrane fusion. In the present study, we performed in vitro reconstitution assays to unravel the molecular mechanisms of EGCG in SNARE-mediated insulin secretory vesicle fusion. Our data show that EGCG efficiently inhibits insulin secretory SNARE-mediated membrane fusion. Mechanistic studies indicated that EGCG blocks the formation of the trans-SNARE complex. Furthermore, calcium/synaptotagmin-7-stimulated fusion kinetics were largely reduced by EGCG, confirming that it is a potential regulator of SNARE-dependent insulin secretion. Our findings suggest that the trans-SNARE complex might be a promising target for controlling SNARE-dependent vesicle fusion.
Collapse
Affiliation(s)
- Min Zhu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life SciencesNanjing Normal UniversityChina
| | - Han Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life SciencesNanjing Normal UniversityChina
| | - Yuting Jiang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life SciencesNanjing Normal UniversityChina
| | - Haijia Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life SciencesNanjing Normal UniversityChina
| | - Yinghui Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life SciencesNanjing Normal UniversityChina
| |
Collapse
|
11
|
Barillaro M, Schuurman M, Wang R. Collagen IV-β1-Integrin Influences INS-1 Cell Insulin Secretion via Enhanced SNARE Protein Expression. Front Cell Dev Biol 2022; 10:894422. [PMID: 35573663 PMCID: PMC9096118 DOI: 10.3389/fcell.2022.894422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/14/2022] [Indexed: 11/18/2022] Open
Abstract
β1-integrin is a key receptor that regulates cell-ECM interactions and is important in maintaining mature beta-cell functions, including insulin secretion. However, there is little reported about the relationship between ECM-β1-integrin interactions and exocytotic proteins involved in glucose-stimulated insulin secretion (GSIS). This study examined the effect of collagen IV-β1-integrin on exocytotic proteins (Munc18-1, Snap25, and Vamp2) involved in insulin secretion using rat insulinoma (INS-1) cell line. Cells cultured on collagen IV (COL IV) had promoted INS-1 cell focal adhesions and GSIS. These cells also displayed changes in levels and localization of β1-integrin associated downstream signals and exocytotic proteins involved in insulin secretion. Antibody blocking of β1-integrin on INS-1 cells cultured on COL IV showed significantly reduced cell adhesion, spreading and insulin secretion along with reduced exocytotic protein levels. Blocking of β1-integrin additionally influenced the cellular localization of exocytotic proteins during the time of GSIS. These results indicate that specific collagen IV-β1-integrin interactions are critical for proper beta-cell insulin secretion.
Collapse
Affiliation(s)
- Malina Barillaro
- Children’s Health Research Institute, London, ON, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - Meg Schuurman
- Children’s Health Research Institute, London, ON, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - Rennian Wang
- Children’s Health Research Institute, London, ON, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
- *Correspondence: Rennian Wang,
| |
Collapse
|
12
|
Merz KE, Hwang J, Zhou C, Veluthakal R, McCown EM, Hamilton A, Oh E, Dai W, Fueger PT, Jiang L, Huss JM, Thurmond DC. Enrichment of the exocytosis protein STX4 in skeletal muscle remediates peripheral insulin resistance and alters mitochondrial dynamics via Drp1. Nat Commun 2022; 13:424. [PMID: 35058456 PMCID: PMC8776765 DOI: 10.1038/s41467-022-28061-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 01/05/2022] [Indexed: 12/15/2022] Open
Abstract
Mitochondrial dysfunction is implicated in skeletal muscle insulin resistance. Syntaxin 4 (STX4) levels are reduced in human diabetic skeletal muscle, and global transgenic enrichment of STX4 expression improves insulin sensitivity in mice. Here, we show that transgenic skeletal muscle-specific STX4 enrichment (skmSTX4tg) in mice reverses established insulin resistance and improves mitochondrial function in the context of diabetogenic stress. Specifically, skmSTX4tg reversed insulin resistance caused by high-fat diet (HFD) without altering body weight or food consumption. Electron microscopy of wild-type mouse muscle revealed STX4 localisation at or proximal to the mitochondrial membrane. STX4 enrichment prevented HFD-induced mitochondrial fragmentation and dysfunction through a mechanism involving STX4-Drp1 interaction and elevated AMPK-mediated phosphorylation at Drp1 S637, which favors fusion. Our findings challenge the dogma that STX4 acts solely at the plasma membrane, revealing that STX4 localises at/proximal to and regulates the function of mitochondria in muscle. These results establish skeletal muscle STX4 enrichment as a candidate therapeutic strategy to reverse peripheral insulin resistance.
Collapse
Affiliation(s)
- Karla E Merz
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA, USA
- Amgen, Thousand Oaks, CA, USA
| | - Jinhee Hwang
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Chunxue Zhou
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Rajakrishnan Veluthakal
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Erika M McCown
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Angelica Hamilton
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Eunjin Oh
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Wenting Dai
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Patrick T Fueger
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
- Comprehensive Metabolic Phenotyping Core, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Lei Jiang
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Janice M Huss
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
- Washington University School of Medicine, St. Louis, MO, USA
| | - Debbie C Thurmond
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA.
| |
Collapse
|
13
|
Mielnicka A, Michaluk P. Exocytosis in Astrocytes. Biomolecules 2021; 11:1367. [PMID: 34572580 PMCID: PMC8471187 DOI: 10.3390/biom11091367] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 12/17/2022] Open
Abstract
Until recently, astrocytes were thought to be a part of a simple "brain glue" providing only a supporting role for neurons. However, the discoveries of the last two decades have proven astrocytes to be dynamic partners participating in brain metabolism and actively influencing communication between neurons. The means of astrocyte-neuron communication are diverse, although regulated exocytosis has received the most attention but also caused the most debate. Similar to most of eukaryotic cells, astrocytes have a complex range of vesicular organelles which can undergo exocytosis as well as intricate molecular mechanisms that regulate this process. In this review, we focus on the components needed for regulated exocytosis to occur and summarise the knowledge about experimental evidence showing its presence in astrocytes.
Collapse
Affiliation(s)
| | - Piotr Michaluk
- BRAINCITY, Laboratory of Neurobiology, The Nencki Institute of Experimental Biology, PAS, 02-093 Warsaw, Poland;
| |
Collapse
|
14
|
Puntman DC, Arora S, Farina M, Toonen RF, Verhage M. Munc18-1 Is Essential for Neuropeptide Secretion in Neurons. J Neurosci 2021; 41:5980-5993. [PMID: 34103363 PMCID: PMC8276746 DOI: 10.1523/jneurosci.3150-20.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 11/21/2022] Open
Abstract
Neuropeptide secretion from dense-core vesicles (DCVs) controls many brain functions. Several components of the DCV exocytosis machinery have recently been identified, but the participation of a SEC1/MUNC18 (SM) protein has remained elusive. Here, we tested the ability of the three exocytic SM proteins expressed in the mammalian brain, MUNC18-1/2/3, to support neuropeptide secretion. We quantified DCV exocytosis at a single vesicle resolution on action potential (AP) train-stimulation in mouse CNS neurons (of unknown sex) using pHluorin-tagged and/or mCherry-tagged neuropeptide Y (NPY) or brain-derived neurotrophic factor (BDNF). Conditional inactivation of Munc18-1 abolished all DCV exocytosis. Expression of MUNC18-1, but not MUNC18-2 or MUNC18-3, supported DCV exocytosis in Munc18-1 null neurons. Heterozygous (HZ) inactivation of Munc18-1, as a model for reduced MUNC18-1 expression, impaired DCV exocytosis, especially during the initial phase of train-stimulation, when the release was maximal. These data show that neurons critically and selectively depend on MUNC18-1 for neuropeptide secretion. Impaired neuropeptide secretion may explain aspects of the behavioral and neurodevelopmental phenotypes that were observed in Munc18-1 HZ mice.SIGNIFICANCE STATEMENT Neuropeptide secretion from dense-core vesicles (DCVs) modulates synaptic transmission, sleep, appetite, cognition and mood. However, the mechanisms of DCV exocytosis are poorly characterized. Here, we identify MUNC18-1 as an essential component for neuropeptide secretion from DCVs. Paralogs MUNC18-2 or MUNC18-3 cannot compensate for MUNC18-1. MUNC18-1 is the first protein identified to be essential for both neuropeptide secretion and synaptic transmission. In heterozygous (HZ) Munc18-1 neurons, that have a 50% reduced MUNC18-1expression and model the human STXBP1 syndrome, DCV exocytosis is impaired, especially during the initial phase of train-stimulation, when the release is maximal. These data show that MUNC18-1 is essential for neuropeptide secretion and that impaired neuropeptide secretion on reduced MUNC18-1expression may contribute to the symptoms of STXBP1 syndrome.
Collapse
Affiliation(s)
- Daniël C Puntman
- Section Functional genomics, Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research, Universitair Medisch Centrum, Amsterdam1081 HV, The Netherlands
| | - Swati Arora
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Margherita Farina
- Section Functional genomics, Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research, Universitair Medisch Centrum, Amsterdam1081 HV, The Netherlands
| | - Ruud F Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Matthijs Verhage
- Section Functional genomics, Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research, Universitair Medisch Centrum, Amsterdam1081 HV, The Netherlands
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam1081 HV, The Netherlands
| |
Collapse
|
15
|
So WY, Liu WN, Teo AKK, Rutter GA, Han W. Paired box 6 programs essential exocytotic genes in the regulation of glucose-stimulated insulin secretion and glucose homeostasis. Sci Transl Med 2021; 13:13/600/eabb1038. [PMID: 34193609 DOI: 10.1126/scitranslmed.abb1038] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 01/25/2021] [Accepted: 05/26/2021] [Indexed: 01/26/2023]
Abstract
The paired box 6 (PAX6) transcription factor is crucial for normal pancreatic islet development and function. Heterozygous mutations of PAX6 are associated with impaired insulin secretion and early-onset diabetes mellitus in humans. However, the molecular mechanism of PAX6 in controlling insulin secretion in human beta cells and its pathophysiological role in type 2 diabetes (T2D) remain ambiguous. We investigated the molecular pathway of PAX6 in the regulation of insulin secretion and the potential therapeutic value of PAX6 in T2D by using human pancreatic beta cell line EndoC-βH1, the db/db mouse model, and primary human pancreatic islets. Through loss- and gain-of-function approaches, we uncovered a mechanism by which PAX6 modulates glucose-stimulated insulin secretion (GSIS) through a cAMP response element-binding protein (CREB)/Munc18-1/2 pathway. Moreover, under diabetic conditions, beta cells and pancreatic islets displayed dampened PAX6/CREB/Munc18-1/2 pathway activity and impaired GSIS, which were reversed by PAX6 replenishment. Adeno-associated virus-mediated PAX6 overexpression in db/db mouse pancreatic beta cells led to a sustained amelioration of glycemic perturbation in vivo but did not affect insulin resistance. Our study highlights the pathophysiological role of PAX6 in T2D-associated beta cell dysfunction in humans and suggests the potential of PAX6 gene transfer in preserving and restoring beta cell function.
Collapse
Affiliation(s)
- Wing Yan So
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research, Singapore 138673, Singapore
| | - Wai Nam Liu
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research, Singapore 138673, Singapore
| | - Adrian Kee Keong Teo
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research, Singapore 138673, Singapore.,Departments of Biochemistry and Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics and Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London W12 0NN, UK
| | - Weiping Han
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research, Singapore 138673, Singapore. .,Center for Neuro-Metabolism and Regeneration Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510700, China
| |
Collapse
|
16
|
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: 4] [Impact Index Per Article: 1.3] [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
|
17
|
Ahn M, Oh E, McCown EM, Wang X, Veluthakal R, Thurmond DC. A requirement for PAK1 to support mitochondrial function and maintain cellular redox balance via electron transport chain proteins to prevent β-cell apoptosis. Metabolism 2021; 115:154431. [PMID: 33181191 PMCID: PMC8123936 DOI: 10.1016/j.metabol.2020.154431] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/04/2020] [Accepted: 11/07/2020] [Indexed: 01/05/2023]
Abstract
OBJECTIVE p21 (Cdc42/Rac1) activated Kinase 1 (PAK1) is a candidate susceptibility factor for type 2 diabetes (T2D). PAK1 is depleted in the islets from T2D donors, compared to control individuals. In addition, whole-body PAK1 knock out (PAK1-KO) in mice worsens the T2D-like effects of high-fat diet. The current study tested the effects of modulating PAK1 levels only in β-cells. MATERIALS/METHODS β-cell-specific inducible PAK1 KO (βPAK1-iKO) mice were generated and used with human β-cells and T2D islets to evaluate β-cell function. RESULTS βPAK1-iKO mice exhibited glucose intolerance and elevated β-cell apoptosis, but without peripheral insulin resistance. β-cells from βPAK-iKO mice also contained fewer mitochondria per cell. At the cellular level, human PAK1-deficient β-cells showed blunted glucose-stimulated insulin secretion and reduced mitochondrial function. Mitochondria from human PAK1-deficient β-cells were deficient in the electron transport chain (ETC) subunits CI, CIII, and CIV; NDUFA12, a CI complex protein, was identified as a novel PAK1 binding partner, and was significantly reduced with PAK1 knockdown. PAK1 knockdown disrupted the NAD+/NADH and NADP+/NADPH ratios, and elevated ROS. An imbalance of the redox state due to mitochondrial dysfunction leads to ER stress in β-cells. PAK1 replenishment in the β-cells of T2D human islets ameliorated levels of ER stress markers. CONCLUSIONS These findings support a protective function for PAK1 in β-cells. The results support a new model whereby the PAK1 in the β-cell plays a required role upstream of mitochondrial function, via maintaining ETC protein levels and averting stress-induced β-cell apoptosis to retain healthy functional β-cell mass.
Collapse
Affiliation(s)
- Miwon Ahn
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute and Beckman Research Institute at the City of Hope, Duarte, CA 91010, United States of America
| | - Eunjin Oh
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute and Beckman Research Institute at the City of Hope, Duarte, CA 91010, United States of America
| | - Erika M McCown
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute and Beckman Research Institute at the City of Hope, Duarte, CA 91010, United States of America
| | - Xin Wang
- Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Rajakrishnan Veluthakal
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute and Beckman Research Institute at the City of Hope, Duarte, CA 91010, United States of America
| | - Debbie C Thurmond
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute and Beckman Research Institute at the City of Hope, Duarte, CA 91010, United States of America.
| |
Collapse
|
18
|
Tang F, Xiao D, Chen L, Gao H, Li X. Role of Munc18-1 in the biological functions and pathogenesis of neurological disorders (Review). Mol Med Rep 2021; 23:198. [PMID: 33495808 PMCID: PMC7821349 DOI: 10.3892/mmr.2021.11837] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 12/30/2020] [Indexed: 11/06/2022] Open
Abstract
The release of neurotransmitters following the fusion of synaptic vesicles and the presynaptic membrane is an important process in the transmission of neuronal information. Syntaxin-binding protein 1 (Munc18-1) is a synaptic fusion protein binding protein, which mainly regulates synaptic vesicle fusion and neurotransmitter release by interacting with soluble N-ethylmaleimide sensitive factor attachment protein receptor. In addition to affecting neurotransmitter transmission, Munc18-1 is also involved in regulating neurosynaptic plasticity, neurodevelopment and neuroendocrine cell release functions (including thyroxine and insulin release). A number of previous studies have demonstrated that Munc18-1 has diverse and vital biological functions, and that its abnormal expression serves an important role in the pathogenesis of a variety of neurological diseases, including epileptic encephalopathy, schizophrenia, autism, Parkinsons disease, Alzheimers disease, multiple sclerosis, Duchennes muscular dystrophy and neuronal ceroid lipofuscinosis. The present review summarizes the function of Munc18-1 and its possible relationship to the pathogenesis of various neurological diseases.
Collapse
Affiliation(s)
- Fajuan Tang
- Department of Emergency, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Dongqiong Xiao
- Department of Emergency, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lin Chen
- Department of Emergency, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hu Gao
- Department of Emergency, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xihong Li
- Department of Emergency, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
19
|
Zhang Z, Li W, Yang G, Lu X, Qi X, Wang S, Cao C, Zhang P, Ren J, Zhao J, Zhang J, Hong S, Tan Y, Burchfield J, Yu Y, Xu T, Yao X, James D, Feng W, Chen Z. CASK modulates the assembly and function of the Mint1/Munc18-1 complex to regulate insulin secretion. Cell Discov 2020; 6:92. [PMID: 33318489 PMCID: PMC7736295 DOI: 10.1038/s41421-020-00216-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/07/2020] [Indexed: 11/09/2022] Open
Abstract
Calcium/calmodulin-dependent protein serine kinase (CASK) is a key player in vesicle transport and release in neurons. However, its precise role, particularly in nonneuronal systems, is incompletely understood. We report that CASK functions as an important regulator of insulin secretion. CASK depletion in mouse islets/β cells substantially reduces insulin secretion and vesicle docking/fusion. CASK forms a ternary complex with Mint1 and Munc18-1, and this event is regulated by glucose stimulation in β cells. The crystal structure of the CASK/Mint1 complex demonstrates that Mint1 exhibits a unique "whip"-like structure that wraps tightly around the CASK-CaMK domain, which contains dual hydrophobic interaction sites. When triggered by CASK binding, Mint1 modulates the assembly of the complex. Further investigation revealed that CASK-Mint1 binding is critical for ternary complex formation, thereby controlling Munc18-1 membrane localization and insulin secretion. Our work illustrates the distinctive molecular basis underlying CASK/Mint1/Munc18-1 complex formation and reveals the importance of the CASK-Mint1-Munc18 signaling axis in insulin secretion.
Collapse
Affiliation(s)
- Zhe Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wei Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Guang Yang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China.,Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Xuefeng Lu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xin Qi
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuting Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Can Cao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Peng Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jinqi Ren
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jiaxu Zhao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Junyi Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Sheng Hong
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yan Tan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - James Burchfield
- Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Yang Yu
- National Center for Protein Science Shanghai, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Tao Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xuebiao Yao
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Laboratory for Physical Sciences at Nanoscale, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - David James
- Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Wei Feng
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China. .,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Zhengjun Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China. .,School of Life Sciences and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
20
|
Kreutzberger AJB, Kiessling V, Doyle CA, Schenk N, Upchurch CM, Elmer-Dixon M, Ward AE, Preobraschenski J, Hussein SS, Tomaka W, Seelheim P, Kattan I, Harris M, Liang B, Kenworthy AK, Desai BN, Leitinger N, Anantharam A, Castle JD, Tamm LK. Distinct insulin granule subpopulations implicated in the secretory pathology of diabetes types 1 and 2. eLife 2020; 9:e62506. [PMID: 33164744 PMCID: PMC7738183 DOI: 10.7554/elife.62506] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Insulin secretion from β-cells is reduced at the onset of type-1 and during type-2 diabetes. Although inflammation and metabolic dysfunction of β-cells elicit secretory defects associated with type-1 or type-2 diabetes, accompanying changes to insulin granules have not been established. To address this, we performed detailed functional analyses of insulin granules purified from cells subjected to model treatments that mimic type-1 and type-2 diabetic conditions and discovered striking shifts in calcium affinities and fusion characteristics. We show that this behavior is correlated with two subpopulations of insulin granules whose relative abundance is differentially shifted depending on diabetic model condition. The two types of granules have different release characteristics, distinct lipid and protein compositions, and package different secretory contents alongside insulin. This complexity of β-cell secretory physiology establishes a direct link between granule subpopulation and type of diabetes and leads to a revised model of secretory changes in the diabetogenic process.
Collapse
Affiliation(s)
- Alex J B Kreutzberger
- Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
- Department for Molecular Physiology and Biological Physics, University of VirginiaCharlottesvilleUnited States
| | - Volker Kiessling
- Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
- Department for Molecular Physiology and Biological Physics, University of VirginiaCharlottesvilleUnited States
| | - Catherine A Doyle
- Department of Pharmacology, University of VirginiaCharlottesvilleUnited States
| | - Noah Schenk
- Department of Pharmacology, University of MichiganAnn ArborUnited States
| | - Clint M Upchurch
- Department of Pharmacology, University of VirginiaCharlottesvilleUnited States
| | - Margaret Elmer-Dixon
- Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
- Department for Molecular Physiology and Biological Physics, University of VirginiaCharlottesvilleUnited States
| | - Amanda E Ward
- Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
- Department for Molecular Physiology and Biological Physics, University of VirginiaCharlottesvilleUnited States
| | - Julia Preobraschenski
- Department of Neurobiology, Max Planck Institute for Biophysical ChemistryGöttingenGermany
- Cluster of Excellence in Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells and Institute for Auditory Neuroscience, University of GöttingenGöttingenGermany
| | - Syed S Hussein
- Department of Microbiology, University of VirginiaCharlottesvilleUnited States
| | - Weronika Tomaka
- Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
- Department for Molecular Physiology and Biological Physics, University of VirginiaCharlottesvilleUnited States
| | - Patrick Seelheim
- Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
- Department for Molecular Physiology and Biological Physics, University of VirginiaCharlottesvilleUnited States
| | - Iman Kattan
- Department of Neurobiology, Max Planck Institute for Biophysical ChemistryGöttingenGermany
| | - Megan Harris
- Department of Cell Biology, University of VirginiaCharlottesvilleUnited States
| | - Binyong Liang
- Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
- Department for Molecular Physiology and Biological Physics, University of VirginiaCharlottesvilleUnited States
| | - Anne K Kenworthy
- Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
- Department for Molecular Physiology and Biological Physics, University of VirginiaCharlottesvilleUnited States
| | - Bimal N Desai
- Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
- Department of Pharmacology, University of VirginiaCharlottesvilleUnited States
| | - Norbert Leitinger
- Department of Pharmacology, University of VirginiaCharlottesvilleUnited States
| | - Arun Anantharam
- Department of Pharmacology, University of MichiganAnn ArborUnited States
| | - J David Castle
- Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
- Department of Cell Biology, University of VirginiaCharlottesvilleUnited States
| | - Lukas K Tamm
- Center for Membrane and Cell Physiology, University of VirginiaCharlottesvilleUnited States
- Department for Molecular Physiology and Biological Physics, University of VirginiaCharlottesvilleUnited States
| |
Collapse
|
21
|
Campbell JR, Martchenko A, Sweeney ME, Maalouf MF, Psichas A, Gribble FM, Reimann F, Brubaker PL. Essential Role of Syntaxin-Binding Protein-1 in the Regulation of Glucagon-Like Peptide-1 Secretion. Endocrinology 2020; 161:5788420. [PMID: 32141504 PMCID: PMC7124137 DOI: 10.1210/endocr/bqaa039] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/28/2020] [Indexed: 12/12/2022]
Abstract
Circadian secretion of the incretin, glucagon-like peptide-1 (GLP-1), correlates with expression of the core clock gene, Bmal1, in the intestinal L-cell. Several SNARE proteins known to be circadian in pancreatic α- and β-cells are also necessary for GLP-1 secretion. However, the role of the accessory SNARE, Syntaxin binding protein-1 (Stxbp1; also known as Munc18-1) in the L-cell is unknown. The aim of this study was to determine whether Stxbp1 is under circadian regulation in the L-cell and its role in the control of GLP-1 secretion. Stxbp1 was highly-enriched in L-cells, and STXBP1 was expressed in a subpopulation of L-cells in mouse and human intestinal sections. Stxbp1 transcripts and protein displayed circadian patterns in mGLUTag L-cells line, while chromatin-immunoprecipitation revealed increased interaction between BMAL1 and Stxbp1 at the peak time-point of the circadian pattern. STXBP1 recruitment to the cytosol and plasma membrane within 30 minutes of L-cell stimulation was also observed at this time-point. Loss of Stxbp1 in vitro and in vivo led to reduced stimulated GLP-1 secretion at the peak time-point of circadian release, and impaired GLP-1 secretion ex vivo. In conclusion, Stxbp1 is a circadian regulated exocytotic protein in the intestinal L-cell that is an essential regulatory component of GLP-1 secretion.
Collapse
Affiliation(s)
| | | | - Maegan E Sweeney
- Departments of Physiology, University of Toronto, Toronto, ON, Canada
| | - Michael F Maalouf
- Departments of Physiology, University of Toronto, Toronto, ON, Canada
| | - Arianna Psichas
- Departments of Medicine, University of Toronto, Toronto, ON, Canada
| | - Fiona M Gribble
- Departments of Medicine, University of Toronto, Toronto, ON, Canada
| | - Frank Reimann
- Departments of Medicine, University of Toronto, Toronto, ON, Canada
| | - Patricia L Brubaker
- Departments of Physiology, University of Toronto, Toronto, ON, Canada
- Wellcome Trust-MRC Institute of Metabolic Science – Metabolic Research Laboratories (IMS-MRL), University of Cambridge, Cambridge, UK
- Correspondence: P.L. Brubaker, Rm. 3366 Medical Sciences Building, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8. E-mail:
| |
Collapse
|
22
|
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: 35] [Impact Index Per Article: 8.8] [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
|
23
|
Biancolin AD, Martchenko A, Mitova E, Gurges P, Michalchyshyn E, Chalmers JA, Doria A, Mychaleckyj JC, Adriaenssens AE, Reimann F, Gribble FM, Gil-Lozano M, Cox BJ, Brubaker PL. The core clock gene, Bmal1, and its downstream target, the SNARE regulatory protein secretagogin, are necessary for circadian secretion of glucagon-like peptide-1. Mol Metab 2020; 31:124-137. [PMID: 31918914 PMCID: PMC6920326 DOI: 10.1016/j.molmet.2019.11.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/24/2019] [Accepted: 11/01/2019] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES The incretin hormone glucagon-like peptide-1 (GLP-1) is secreted from intestinal L-cells upon nutrient intake. While recent evidence has shown that GLP-1 is released in a circadian manner in rats, whether this occurs in mice and if this pattern is regulated by the circadian clock remain to be elucidated. Furthermore, although circadian GLP-1 secretion parallels expression of the core clock gene Bmal1, the link between the two remains largely unknown. Secretagogin (Scgn) is an exocytotic SNARE regulatory protein that demonstrates circadian expression and is essential for insulin secretion from β-cells. The objective of the current study was to establish the necessity of the core clock gene Bmal1 and the SNARE protein SCGN as essential regulators of circadian GLP-1 secretion. METHODS Oral glucose tolerance tests were conducted at different times of the day on 4-hour fasted C57BL/6J, Bmal1 wild-type, and Bmal1 knockout mice. Mass spectrometry, RNA-seq, qRT-PCR and/or microarray analyses, and immunostaining were conducted on murine (m) and human (h) primary L-cells and mGLUTag and hNCI-H716 L-cell lines. At peak and trough GLP-1 secretory time points, the mGLUTag cells were co-stained for SCGN and a membrane-marker, ChIP was used to analyze BMAL1 binding sites in the Scgn promoter, protein interaction with SCGN was tested by co-immunoprecipitation, and siRNA was used to knockdown Scgn for GLP-1 secretion assay. RESULTS C57BL/6J mice displayed a circadian rhythm in GLP-1 secretion that peaked at the onset of their feeding period. Rhythmic GLP-1 release was impaired in Bmal1 knockout (KO) mice as compared to wild-type controls at the peak (p < 0.05) but not at the trough secretory time point. Microarray identified SNARE and transport vesicle pathways as highly upregulated in mGLUTag L-cells at the peak time point of GLP-1 secretion (p < 0.001). Mass spectrometry revealed that SCGN was also increased at this time (p < 0.001), while RNA-seq, qRT-PCR, and immunostaining demonstrated Scgn expression in all human and murine primary L-cells and cell lines. The mGLUTag and hNCI-H716 L-cells exhibited circadian rhythms in Scgn expression (p < 0.001). The ChIP analysis demonstrated increased binding of BMAL1 only at the peak of Scgn expression (p < 0.01). Immunocytochemistry showed the translocation of SCGN to the cell membrane after stimulation at the peak time point only (p < 0.05), while CoIP showed that SCGN was pulled down with SNAP25 and β-actin, but only the latter interaction was time-dependent (p < 0.05). Finally, Scgn siRNA-treated cells demonstrated significantly blunted GLP-1 secretion (p < 0.01) in response to stimulation at the peak time point only. CONCLUSIONS These data demonstrate, for the first time, that mice display a circadian pattern in GLP-1 secretion, which is impaired in Bmal1 knockout mice, and that Bmal1 regulation of Scgn expression plays an essential role in the circadian release of the incretin hormone GLP-1.
Collapse
Affiliation(s)
| | | | - Emilia Mitova
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Patrick Gurges
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | | | | | - Alessandro Doria
- Department of Medicine, Harvard Medical School, Boston, MA, USA; Research Division, Joslin Diabetes Center, Boston, MA, USA
| | - Josyf C Mychaleckyj
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Alice E Adriaenssens
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Frank Reimann
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Fiona M Gribble
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Manuel Gil-Lozano
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Brian J Cox
- Department of Physiology, University of Toronto, Toronto, ON, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada
| | - Patricia L Brubaker
- Department of Physiology, University of Toronto, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
24
|
In vitro fusion of single synaptic and dense core vesicles reproduces key physiological properties. Nat Commun 2019; 10:3904. [PMID: 31467284 PMCID: PMC6715626 DOI: 10.1038/s41467-019-11873-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 08/02/2019] [Indexed: 12/29/2022] Open
Abstract
Regulated exocytosis of synaptic vesicles is substantially faster than of endocrine dense core vesicles despite similar molecular machineries. The reasons for this difference are unknown and could be due to different regulatory proteins, different spatial arrangements, different vesicle sizes, or other factors. To address these questions, we take a reconstitution approach and compare regulated SNARE-mediated fusion of purified synaptic and dense core chromaffin and insulin vesicles using a single vesicle-supported membrane fusion assay. In all cases, Munc18 and complexin are required to restrict fusion in the absence of calcium. Calcium triggers fusion of all docked vesicles. Munc13 (C1C2MUN domain) is required for synaptic and enhanced insulin vesicle fusion, but not for chromaffin vesicles, correlating inversely with the presence of CAPS protein on purified vesicles. Striking disparities in calcium-triggered fusion rates are observed, increasing with curvature with time constants 0.23 s (synaptic vesicles), 3.3 s (chromaffin vesicles), and 9.1 s (insulin vesicles) and correlating with rate differences in cells.
Collapse
|
25
|
Jaramillo AM, Piccotti L, Velasco WV, Delgado ASH, Azzegagh Z, Chung F, Nazeer U, Farooq J, Brenner J, Parker-Thornburg J, Scott BL, Evans CM, Adachi R, Burns AR, Kreda SM, Tuvim MJ, Dickey BF. Different Munc18 proteins mediate baseline and stimulated airway mucin secretion. JCI Insight 2019; 4:124815. [PMID: 30721150 PMCID: PMC6483006 DOI: 10.1172/jci.insight.124815] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 01/31/2019] [Indexed: 12/15/2022] Open
Abstract
Airway mucin secretion is necessary for ciliary clearance of inhaled particles and pathogens but can be detrimental in pathologies such as asthma and cystic fibrosis. Exocytosis in mammals requires a Munc18 scaffolding protein, and airway secretory cells express all 3 Munc18 isoforms. Using conditional airway epithelial cell-deletant mice, we found that Munc18a has the major role in baseline mucin secretion, Munc18b has the major role in stimulated mucin secretion, and Munc18c does not function in mucin secretion. In an allergic asthma model, Munc18b deletion reduced airway mucus occlusion and airflow resistance. In a cystic fibrosis model, Munc18b deletion reduced airway mucus occlusion and emphysema. Munc18b deficiency in the airway epithelium did not result in any abnormalities of lung structure, particle clearance, inflammation, or bacterial infection. Our results show that regulated secretion in a polarized epithelial cell may involve more than one exocytic machine at the apical plasma membrane and that the protective roles of mucin secretion can be preserved while therapeutically targeting its pathologic roles.
Collapse
Affiliation(s)
- Ana M. Jaramillo
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Institute of Bioscience and Technology, Texas A&M University Health Science Center, Houston, Texas, USA
| | - Lucia Piccotti
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Walter V. Velasco
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Zoulikha Azzegagh
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Felicity Chung
- Marsico Lung Institute/Cystic Fibrosis Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Usman Nazeer
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Junaid Farooq
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Josh Brenner
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jan Parker-Thornburg
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Brenton L. Scott
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Christopher M. Evans
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Roberto Adachi
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Alan R. Burns
- College of Optometry, University of Houston, Houston, Texas, USA
| | - Silvia M. Kreda
- Marsico Lung Institute/Cystic Fibrosis Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michael J. Tuvim
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Burton F. Dickey
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
26
|
Cardenas EI, Gonzalez R, Breaux K, Da Q, Gutierrez BA, Ramos MA, Cardenas RA, Burns AR, Rumbaut RE, Adachi R. Munc18-2, but not Munc18-1 or Munc18-3, regulates platelet exocytosis, hemostasis, and thrombosis. J Biol Chem 2019; 294:4784-4792. [PMID: 30696774 DOI: 10.1074/jbc.ra118.006922] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/24/2019] [Indexed: 12/17/2022] Open
Abstract
Platelet degranulation, a form of regulated exocytosis, is crucial for hemostasis and thrombosis. Exocytosis in platelets is mediated by SNARE proteins, and in most mammalian cells this process is controlled by Munc18 (mammalian homolog of Caenorhabditis elegans uncoordinated gene 18) proteins. Platelets express all Munc18 paralogs (Munc18-1, -2, and -3), but their roles in platelet secretion and function have not been fully characterized. Using Munc18-1, -2, and -3 conditional knockout mice, here we deleted expression of these proteins in platelets and assessed granule exocytosis. We measured products secreted by each type of platelet granule and analyzed EM platelet profiles by design-based stereology. We observed that the removal of Munc18-2 ablates the release of alpha, dense, and lysosomal granules from platelets, but we found no exocytic role for Munc18-1 or -3 in platelets. In vitro, Munc18-2-deficient platelets exhibited defective aggregation at low doses of collagen and impaired thrombus formation under shear stress. In vivo, megakaryocyte-specific Munc18-2 conditional knockout mice had a severe hemostatic defect and prolonged arterial and venous bleeding times. They were also protected against arterial thrombosis in a chemically induced model of arterial injury. Taken together, our results indicate that Munc18-2, but not Munc18-1 or Munc18-3, is essential for regulated exocytosis in platelets and platelet participation in thrombosis and hemostasis.
Collapse
Affiliation(s)
- Eduardo I Cardenas
- From the Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030.,the Escuela de Ingenieria y Ciencias, Instituto Tecnologico de Monterrey, Monterrey, Nuevo Leon 64849, Mexico
| | - Ricardo Gonzalez
- From the Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030.,the Escuela de Ingenieria y Ciencias, Instituto Tecnologico de Monterrey, Monterrey, Nuevo Leon 64849, Mexico
| | - Keegan Breaux
- From the Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Qi Da
- the Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas 77030.,the Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| | - Berenice A Gutierrez
- From the Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030.,the Escuela de Ingenieria y Ciencias, Instituto Tecnologico de Monterrey, Monterrey, Nuevo Leon 64849, Mexico
| | - Marco A Ramos
- From the Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Rodolfo A Cardenas
- From the Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030.,the Escuela de Medicina y Ciencias de la Salud, Instituto Tecnologico de Monterrey, Monterrey, Nuevo León 64710, México, and
| | - Alan R Burns
- the College of Optometry, University of Houston, Houston, Texas 77204
| | - Rolando E Rumbaut
- the Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas 77030.,the Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| | - Roberto Adachi
- From the Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030,
| |
Collapse
|
27
|
Oh E, Ahn M, Afelik S, Becker TC, Roep BO, Thurmond DC. Syntaxin 4 Expression in Pancreatic β-Cells Promotes Islet Function and Protects Functional β-Cell Mass. Diabetes 2018; 67:2626-2639. [PMID: 30305365 PMCID: PMC6245223 DOI: 10.2337/db18-0259] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 09/25/2018] [Indexed: 02/06/2023]
Abstract
Syntaxin 4 (Stx4) enrichment in human and mouse islet grafts improves the success of transplants in reversing streptozotocin (STZ)-induced diabetes in mice, although the underlying molecular mechanisms remain elusive. Toward a further understanding of this, human islets and inducible transgenic mice that selectively overexpress Stx4 in islet β-cells (βTG-Stx4) were challenged with proinflammatory stressors in vitro and in vivo. Remarkably, βTG-Stx4 mice resisted the loss of β-cell mass and the glucose intolerance that multiple low doses of STZ induce. Under standard conditions, glucose tolerance was enhanced and mice maintained normal fasting glycemia and insulinemia. Conversely, Stx4 heterozygous knockout mice succumbed rapidly to STZ-induced glucose intolerance compared with their wild-type littermates. Human islet β-cells overexpressing Stx4 exhibited enhanced insulin secretory capability; resilience against proinflammatory cytokine-induced apoptosis; and reduced expression of the CXCL9, CXCL10, and CXCL11 genes coordinate with decreased activation/nuclear localization of nuclear factor-κB. Finding ways to boost Stx4 expression presents a novel potential therapeutic avenue for promoting islet function and preserving β-cell mass.
Collapse
Affiliation(s)
- Eunjin Oh
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA
| | - Miwon Ahn
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA
| | - Solomon Afelik
- Department of Surgery/Division of Transplantation, University of Illinois at Chicago, Chicago, IL
| | - Thomas C Becker
- Department of Internal Medicine, Duke Molecular Physiology Institute, Duke University, Durham, NC
| | - Bart O Roep
- Department of Diabetes Immunology, Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA
| | - Debbie C Thurmond
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA
| |
Collapse
|
28
|
Marré ML, Piganelli JD, James EA. Protecting functional β cells with a therapeutic peptide. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:372. [PMID: 30370299 DOI: 10.21037/atm.2018.07.26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Meghan L Marré
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jon D Piganelli
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eddie A James
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| |
Collapse
|
29
|
Aslamy A, Oh E, Olson EM, Zhang J, Ahn M, Moin ASM, Tunduguru R, Salunkhe VA, Veluthakal R, Thurmond DC. Doc2b Protects β-Cells Against Inflammatory Damage and Enhances Function. Diabetes 2018; 67:1332-1344. [PMID: 29661782 PMCID: PMC6014558 DOI: 10.2337/db17-1352] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 04/09/2018] [Indexed: 12/12/2022]
Abstract
Loss of functional β-cell mass is an early feature of type 1 diabetes. To release insulin, β-cells require soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes, as well as SNARE complex regulatory proteins like double C2 domain-containing protein β (Doc2b). We hypothesized that Doc2b deficiency or overabundance may confer susceptibility or protection, respectively, to the functional β-cell mass. Indeed, Doc2b+/- knockout mice show an unusually severe response to multiple-low-dose streptozotocin (MLD-STZ), resulting in more apoptotic β-cells and a smaller β-cell mass. In addition, inducible β-cell-specific Doc2b-overexpressing transgenic (βDoc2b-dTg) mice show improved glucose tolerance and resist MLD-STZ-induced disruption of glucose tolerance, fasting hyperglycemia, β-cell apoptosis, and loss of β-cell mass. Mechanistically, Doc2b enrichment enhances glucose-stimulated insulin secretion (GSIS) and SNARE activation and prevents the appearance of apoptotic markers in response to cytokine stress and thapsigargin. Furthermore, expression of a peptide containing the Doc2b tandem C2A and C2B domains is sufficient to confer the beneficial effects of Doc2b enrichment on GSIS, SNARE activation, and apoptosis. These studies demonstrate that Doc2b enrichment in the β-cell protects against diabetogenic and proapoptotic stress. Furthermore, they identify a Doc2b peptide that confers the beneficial effects of Doc2b and may be a therapeutic candidate for protecting functional β-cell mass.
Collapse
Affiliation(s)
- Arianne Aslamy
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolic Research Institute, Beckman Research Institute of City of Hope, Duarte, CA
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
| | - Eunjin Oh
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolic Research Institute, Beckman Research Institute of City of Hope, Duarte, CA
| | - Erika M Olson
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolic Research Institute, Beckman Research Institute of City of Hope, Duarte, CA
| | - Jing Zhang
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolic Research Institute, Beckman Research Institute of City of Hope, Duarte, CA
| | - Miwon Ahn
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolic Research Institute, Beckman Research Institute of City of Hope, Duarte, CA
| | - Abu Saleh Md Moin
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolic Research Institute, Beckman Research Institute of City of Hope, Duarte, CA
| | - Ragadeepthi Tunduguru
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolic Research Institute, Beckman Research Institute of City of Hope, Duarte, CA
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolic Research Institute, Beckman Research Institute of City of Hope, Duarte, CA
| | - Vishal A Salunkhe
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolic Research Institute, Beckman Research Institute of City of Hope, Duarte, CA
| | - Rajakrishnan Veluthakal
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolic Research Institute, Beckman Research Institute of City of Hope, Duarte, CA
| | - Debbie C Thurmond
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolic Research Institute, Beckman Research Institute of City of Hope, Duarte, CA
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
30
|
Zhao D, Ma L, Shen C, Li D, Cheng W, Shang Y, Liu Z, Wang X, Yin K. Long-lasting Glucagon-like Peptide 1 Analogue Exendin-4 Ameliorates the Secretory and Synthetic Function of Islets Isolated From Severely Scalded Rats. J Burn Care Res 2018; 39:545-554. [PMID: 29579298 DOI: 10.1093/jbcr/irx014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Dongxu Zhao
- Department of Burn and Plastic Surgery, Burns Institute, the First Affiliated Hospital of General Hospital of PLA, Beijing, China
| | - Li Ma
- Department of Burn and Plastic Surgery, Burns Institute, the First Affiliated Hospital of General Hospital of PLA, Beijing, China
| | - Chuanan Shen
- Department of Burn and Plastic Surgery, Burns Institute, the First Affiliated Hospital of General Hospital of PLA, Beijing, China
| | - Dawei Li
- Department of Burn and Plastic Surgery, Burns Institute, the First Affiliated Hospital of General Hospital of PLA, Beijing, China
| | - Wenfeng Cheng
- Department of Burn and Plastic Surgery, Burns Institute, the First Affiliated Hospital of General Hospital of PLA, Beijing, China
| | - Yuru Shang
- Department of Burn and Plastic Surgery, Burns Institute, the First Affiliated Hospital of General Hospital of PLA, Beijing, China
| | - Zhaoxing Liu
- Department of Burn and Plastic Surgery, Burns Institute, the First Affiliated Hospital of General Hospital of PLA, Beijing, China
| | - Xin Wang
- Department of Burn and Plastic Surgery, Burns Institute, the First Affiliated Hospital of General Hospital of PLA, Beijing, China
| | - Kai Yin
- Department of Burn and Plastic Surgery, Burns Institute, the First Affiliated Hospital of General Hospital of PLA, Beijing, China
| |
Collapse
|
31
|
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: 2.1] [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
|
32
|
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: 1] [Impact Index Per Article: 0.1] [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
|
33
|
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: 47] [Impact Index Per Article: 6.7] [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
|
34
|
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: 24] [Impact Index Per Article: 3.4] [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
|
35
|
Molecular regulation of insulin granule biogenesis and exocytosis. Biochem J 2017; 473:2737-56. [PMID: 27621482 DOI: 10.1042/bcj20160291] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 04/19/2016] [Indexed: 12/15/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disorder characterized by hyperglycemia, insulin resistance and hyperinsulinemia in early disease stages but a relative insulin insufficiency in later stages. Insulin, a peptide hormone, is produced in and secreted from pancreatic β-cells following elevated blood glucose levels. Upon its release, insulin induces the removal of excessive exogenous glucose from the bloodstream primarily by stimulating glucose uptake into insulin-dependent tissues as well as promoting hepatic glycogenesis. Given the increasing prevalence of T2DM worldwide, elucidating the underlying mechanisms and identifying the various players involved in the synthesis and exocytosis of insulin from β-cells is of utmost importance. This review summarizes our current understanding of the route insulin takes through the cell after its synthesis in the endoplasmic reticulum as well as our knowledge of the highly elaborate network that controls insulin release from the β-cell. This network harbors potential targets for anti-diabetic drugs and is regulated by signaling cascades from several endocrine systems.
Collapse
|
36
|
Zhu D, Xie L, Kang Y, Dolai S, Bondo Hansen J, Qin T, Xie H, Liang T, Rubin DC, Osborne L, Gaisano HY. Syntaxin 2 Acts as Inhibitory SNARE for Insulin Granule Exocytosis. Diabetes 2017; 66:948-959. [PMID: 28115395 PMCID: PMC5860373 DOI: 10.2337/db16-0636] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 12/24/2016] [Indexed: 01/03/2023]
Abstract
Of the four syntaxins specialized for exocytosis, syntaxin (Syn)-2 is the least understood. In this study, we used Syn-2/epimorphin knockout mice to examine the role of Syn-2 in insulin secretory granule (SG) exocytosis. Unexpectedly, Syn-2 knockout mice exhibited paradoxical superior glucose homeostasis resulting from an enhanced insulin secretion. This was confirmed in vitro by pancreatic islet perifusion showing an amplified biphasic glucose-stimulated insulin secretion arising from an increase in size of the readily releasable pool of insulin SGs and enhanced SG pool refilling. The increase in insulin exocytosis was attributed mainly to an enhanced recruitment of the larger pool of newcomer SGs that undergoes no residence time on plasma membrane before fusion and, to a lesser extent, also the predocked SGs. Consistently, Syn-2 depletion resulted in a stimulation-induced increase in abundance of exocytotic complexes we previously demonstrated as mediating the fusion of newcomer SGs (Syn-3/VAMP8/SNAP25/Munc18b) and predocked SGs (Syn-1A/VAMP2/SNAP25/Muncn18a). This work is the first to show in mammals that Syn-2 could function as an inhibitory SNARE protein that, when relieved, could promote exocytosis in pancreatic islet β-cells. Thus, Syn-2 may serve as a potential target to treat diabetes.
Collapse
Affiliation(s)
- Dan Zhu
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Li Xie
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Youhou Kang
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Subhankar Dolai
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Tairan Qin
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Huanli Xie
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Tao Liang
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Deborah C Rubin
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Lucy Osborne
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Herbert Y Gaisano
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
37
|
Aslamy A, Thurmond DC. Exocytosis proteins as novel targets for diabetes prevention and/or remediation? Am J Physiol Regul Integr Comp Physiol 2017; 312:R739-R752. [PMID: 28356294 DOI: 10.1152/ajpregu.00002.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/24/2017] [Accepted: 03/24/2017] [Indexed: 12/17/2022]
Abstract
Diabetes remains one of the leading causes of morbidity and mortality worldwide, affecting an estimated 422 million adults. In the US, it is predicted that one in every three children born as of 2000 will suffer from diabetes in their lifetime. Type 2 diabetes results from combinatorial defects in pancreatic β-cell glucose-stimulated insulin secretion and in peripheral glucose uptake. Both processes, insulin secretion and glucose uptake, are mediated by exocytosis proteins, SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) complexes, Sec1/Munc18 (SM), and double C2-domain protein B (DOC2B). Increasing evidence links deficiencies in these exocytosis proteins to diabetes in rodents and humans. Given this, emerging studies aimed at restoring and/or enhancing cellular levels of certain exocytosis proteins point to promising outcomes in maintaining functional β-cell mass and enhancing insulin sensitivity. In doing so, new evidence also shows that enhancing exocytosis protein levels may promote health span and longevity and may also harbor anti-cancer and anti-Alzheimer's disease capabilities. Herein, we present a comprehensive review of the described capabilities of certain exocytosis proteins and how these might be targeted for improving metabolic dysregulation.
Collapse
Affiliation(s)
- Arianne Aslamy
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana; and
| | - Debbie C Thurmond
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana; and .,Department of Molecular and Cellular Endocrinology, Beckman Research Institute of City of Hope, Duarte, California
| |
Collapse
|
38
|
Eliasson L, Esguerra JLS, Wendt A. Lessons from basic pancreatic beta cell research in type-2 diabetes and vascular complications. Diabetol Int 2017; 8:139-152. [PMID: 30603317 DOI: 10.1007/s13340-017-0304-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 01/08/2017] [Indexed: 12/14/2022]
Abstract
The changes in life-style with increased access of food and reduced physical activity have resulted in the global epidemic of obesity. Consequently, individuals with type 2 diabetes and cardiovascular disease have also escalated. A central organ in the development of diabetes is the pancreas, and more specifically the pancreatic beta cells within the islets of Langerhans. Beta cells have been assigned the important task of secreting insulin when blood glucose is increased to lower the glucose level. An early sign of diabetes pathogenesis is lack of first phase insulin response and reduced second phase secretion. In this review, which is based on the foreign investigator award lecture given at the JSDC meeting in Sendai in October 2016, we discuss a possible cellular explanation for the reduced first phase insulin response and how this can be influenced by lipids. Moreover, since patients with cardiovascular disease and high levels of cholesterol are often treated with statins, we summarize recent data regarding effects on statins on glucose homeostasis and insulin secretion. Finally, we suggest microRNAs (miRNAs) as central players in the adjustment of beta cell function during the development of diabetes. We specifically discuss miRNAs regarding their involvement in insulin secretion regulation, differential expression in type 2 diabetes, and potential as biomarkers for prediction of diabetes and cardiovascular complications.
Collapse
Affiliation(s)
- Lena Eliasson
- Islet Cell Exocytosis, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Clinical Research Centre, SUS 91-11, Box 50332, 202 13 Malmö, Sweden
| | - Jonathan Lou S Esguerra
- Islet Cell Exocytosis, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Clinical Research Centre, SUS 91-11, Box 50332, 202 13 Malmö, Sweden
| | - Anna Wendt
- Islet Cell Exocytosis, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Clinical Research Centre, SUS 91-11, Box 50332, 202 13 Malmö, Sweden
| |
Collapse
|
39
|
Bohmbach K, Schwarz MK, Schoch S, Henneberger C. The structural and functional evidence for vesicular release from astrocytes in situ. Brain Res Bull 2017; 136:65-75. [PMID: 28122264 DOI: 10.1016/j.brainresbull.2017.01.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 01/17/2017] [Accepted: 01/19/2017] [Indexed: 12/31/2022]
Abstract
The concept of the tripartite synapse states that bi-directional signalling between perisynaptic astrocyte processes, presynaptic axonal boutons and postsynaptic neuronal structures defines the properties of synaptic information processing. Ca2+-dependent vesicular release from astrocytes, as one of the mechanisms of astrocyte-neuron communication, has attracted particular attention but has also been the subject of intense debate. In neurons, regulated vesicular release is a strongly coordinated process. It requires a complex release machinery comprised of many individual components ranging from vesicular neurotransmitter transporters and soluble NSF attachment protein receptors (SNARE) proteins to Ca2+-sensors and the proteins that spatially and temporally control exocytosis of synaptic vesicles. If astrocytes employ similar mechanisms to release neurotransmitters is less well understood. The aim of this review is therefore to discuss recent experimental evidence that sheds light on the central structural components responsible for vesicular release from astrocytes in situ.
Collapse
Affiliation(s)
- Kirsten Bohmbach
- Institute of Cellular Neurosciences, University of Bonn Medical School, Bonn, Germany.
| | - Martin K Schwarz
- Department of Epileptology, University of Bonn Medical School, Bonn, Germany
| | - Susanne Schoch
- Institute of Neuropathology, University of Bonn Medical School, Bonn, Germany
| | - Christian Henneberger
- Institute of Cellular Neurosciences, University of Bonn Medical School, Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; Institute of Neurology, University College London, London, United Kingdom.
| |
Collapse
|
40
|
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: 2.3] [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
|
41
|
Osteopontin Affects Insulin Vesicle Localization and Ca2+ Homeostasis in Pancreatic Beta Cells from Female Mice. PLoS One 2017; 12:e0170498. [PMID: 28107503 PMCID: PMC5249066 DOI: 10.1371/journal.pone.0170498] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 12/15/2016] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetic patients suffer from insulin resistance and reduced insulin secretion. Osteopontin (OPN), a versatile protein expressed in several tissues throughout the body including the islets of Langerhans, has previously been implicated in the development of insulin resistance. Here we have investigated the role of OPN in insulin secretion using an OPN knock out mouse model (OPN-/-). Ultra-structural analyzes of islets from OPN-/- and WT mice indicated weaker cell-cell connections between the islet cells in the OPN-/- mouse compared to WT. Analysis of the insulin granule distribution in the beta cells showed that although OPN-/- and WT beta cells have the same number of insulin granules OPN-/- beta cells have significantly fewer docked granules. Both OPN-/- and WT islets displayed synchronized Ca2+ oscillations indicative of an intact beta cell communication. OPN-/- islets displayed higher intracellular Ca2+ concentrations when stimulated with 16.7 mM glucose than WT islets and the initial dip upon elevated glucose concentrations (which is associated with Ca2+ uptake into ER) was significantly lower in these islets. Glucose-induced insulin secretion was similar in OPN-/- and WT islets. Likewise, non-fasted blood glucose levels were the same in both groups. In summary, deletion of OPN results in several minor beta-cell defects that can be compensated for in a healthy system.
Collapse
|
42
|
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: 3.0] [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
|
43
|
Ahn M, Yoder SM, Wang Z, Oh E, Ramalingam L, Tunduguru R, Thurmond DC. The p21-activated kinase (PAK1) is involved in diet-induced beta cell mass expansion and survival in mice and human islets. Diabetologia 2016; 59:2145-55. [PMID: 27394663 PMCID: PMC5266538 DOI: 10.1007/s00125-016-4042-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/10/2016] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS Human islets from type 2 diabetic donors are reportedly 80% deficient in the p21 (Cdc42/Rac)-activated kinase, PAK1. PAK1 is implicated in beta cell function and maintenance of beta cell mass. We questioned the mechanism(s) by which PAK1 deficiency potentially contributes to increased susceptibility to type 2 diabetes. METHODS Non-diabetic human islets and INS 832/13 beta cells cultured under diabetogenic conditions (i.e. with specific cytokines or under glucolipotoxic [GLT] conditions) were evaluated for changes to PAK1 signalling. Combined effects of PAK1 deficiency with GLT stress were assessed using classic knockout (Pak1 (-/-) ) mice fed a 45% energy from fat/palmitate-based, 'western' diet (WD). INS 832/13 cells overexpressing or depleted of PAK1 were also assessed for apoptosis and signalling changes. RESULTS Exposure of non-diabetic human islets to diabetic stressors attenuated PAK1 protein levels, concurrent with increased caspase 3 cleavage. WD-fed Pak1 knockout mice exhibited fasting hyperglycaemia and severe glucose intolerance. These mice also failed to mount an insulin secretory response following acute glucose challenge, coinciding with a 43% loss of beta cell mass when compared with WD-fed wild-type mice. Pak1 knockout mice had fewer total beta cells per islet, coincident with decreased beta cell proliferation. In INS 832/13 beta cells, PAK1 deficiency combined with GLT exposure heightened beta cell death relative to either condition alone; PAK1 deficiency resulted in decreased extracellular signal-related kinase (ERK) and B cell lymphoma 2 (Bcl2) phosphorylation levels. Conversely, PAK1 overexpression prevented GLT-induced cell death. CONCLUSIONS/INTERPRETATION These findings suggest that PAK1 deficiency may underlie an increased diabetic susceptibility. Discovery of ways to remediate glycaemic dysregulation via altering PAK1 or its downstream effectors offers promising opportunities for disease intervention.
Collapse
Affiliation(s)
- Miwon Ahn
- Department of Molecular & Cellular Endocrinology, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Stephanie M Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Zhanxiang Wang
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Eunjin Oh
- Department of Molecular & Cellular Endocrinology, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Latha Ramalingam
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ragadeepthi Tunduguru
- Department of Molecular & Cellular Endocrinology, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Debbie C Thurmond
- Department of Molecular & Cellular Endocrinology, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA.
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
44
|
Xie L, Zhu D, Dolai S, Liang T, Qin T, Kang Y, Xie H, Huang YC, Gaisano HY. Syntaxin-4 mediates exocytosis of pre-docked and newcomer insulin granules underlying biphasic glucose-stimulated insulin secretion in human pancreatic beta cells. Diabetologia 2015; 58:1250-9. [PMID: 25762204 DOI: 10.1007/s00125-015-3545-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 02/03/2015] [Indexed: 10/23/2022]
Abstract
AIMS/HYPOTHESIS Of the four exocytotic syntaxins (Syns), much is now known about the role of Syn-1A (pre-docked secretory granules [SGs]) and Syn-3 (newcomer SGs) in insulin exocytosis. Some work was reported on Syn-4's role in biphasic glucose-stimulated insulin secretion (GSIS), but its precise role in insulin SG exocytosis remains unclear. In this paper we examine this role in human beta cells. METHODS Endogenous function of Syn-4 in human islets was assessed by knocking down its expression with lentiviral single hairpin RNA (lenti-shRNA)-RFP. Biphasic GSIS was determined by islet perifusion assay. Single-cell analysis of exocytosis of red fluorescent protein (RFP)-positive beta cells (exhibiting near-total depletion of Syn-4) was by patch clamp capacitance measurements (Cm) and total internal reflection fluorescence microscopy (TIRFM), the latter to further assess single SG behaviour. Co-immunoprecipitations were conducted on INS-1 cells to assess exocytotic complexes. RESULTS Syn-4 knockdown (KD) of 77% in human islets caused a concomitant reduction in cognate Munc18c expression (46%) without affecting expression of other exocytotic proteins; this resulted in reduction of GSIS in the first phase (by 42%) and the second phase (by 40%). Cm of RFP-tagged Syn-4-KD beta cells showed severe inhibition in the readily releasable pool (by 71%) and mobilisation from reserve pools (by 63%). TIRFM showed that Syn-4-KD-induced inhibition of first-phase GSIS was attributed to reduction in exocytosis of both pre-docked and newcomer SGs (which undergo minimal residence or docking time at the plasma membrane before fusion). Second-phase inhibition was attributed to reduction in newcomer SGs. Stx-4 co-immunoprecipitated Munc18c, VAMP2 and VAMP8, suggesting that these exocytotic complexes may be involved in exocytosis of pre-docked and newcomer SGs. CONCLUSIONS/INTERPRETATION Syn-4 is involved in distinct molecular machineries that influence exocytosis of both pre-docked and newcomer SGs in a manner functionally redundant to Syn-1A and Syn-3, respectively; this underlies Syn-4's role in mediating portions of first-phase and second-phase GSIS.
Collapse
Affiliation(s)
- Li Xie
- Department of Medicine, Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, Canada, M5S 1A8
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kayton NS, Poffenberger G, Henske J, Dai C, Thompson C, Aramandla R, Shostak A, Nicholson W, Brissova M, Bush WS, Powers AC. Human islet preparations distributed for research exhibit a variety of insulin-secretory profiles. Am J Physiol Endocrinol Metab 2015; 308:E592-602. [PMID: 25648831 PMCID: PMC4385877 DOI: 10.1152/ajpendo.00437.2014] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 01/30/2015] [Indexed: 12/21/2022]
Abstract
Human islet research is providing new insights into human islet biology and diabetes, using islets isolated at multiple US centers from donors with varying characteristics. This creates challenges for understanding, interpreting, and integrating research findings from the many laboratories that use these islets. In what is, to our knowledge, the first standardized assessment of human islet preparations from multiple isolation centers, we measured insulin secretion from 202 preparations isolated at 15 centers over 11 years and noted five distinct patterns of insulin secretion. Approximately three quarters were appropriately responsive to stimuli, but one quarter were dysfunctional, with unstable basal insulin secretion and/or an impairment in stimulated insulin secretion. Importantly, the patterns of insulin secretion by responsive human islet preparations (stable Baseline and Fold stimulation of insulin secretion) isolated at different centers were similar and improved slightly over the years studied. When all preparations studied were considered, basal and stimulated insulin secretion did not correlate with isolation center, biological differences of the islet donor, or differences in isolation, such as Cold Ischemia Time. Dysfunctional islet preparations could not be predicted from the information provided by the isolation center and had altered expression of genes encoding components of the glucose-sensing pathway, but not of insulin production or cell death. These results indicate that insulin secretion by most preparations from multiple centers is similar but that in vitro responsiveness of human islets cannot be predicted, necessitating preexperimental human islet assessment. These results should be considered when one is designing, interpreting, and integrating experiments using human islets.
Collapse
Affiliation(s)
- Nora S Kayton
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Gregory Poffenberger
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joseph Henske
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Chunhua Dai
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Courtney Thompson
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Radhika Aramandla
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Alena Shostak
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Wendell Nicholson
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Marcela Brissova
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - William S Bush
- Center for Human Genetics Research, Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Alvin C Powers
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
| |
Collapse
|
46
|
Doc2b serves as a scaffolding platform for concurrent binding of multiple Munc18 isoforms in pancreatic islet β-cells. Biochem J 2015; 464:251-8. [PMID: 25190515 DOI: 10.1042/bj20140845] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Biphasic glucose-stimulated insulin secretion (GSIS) from pancreatic β-cells involves soluble N-ethylmaleimide-sensitive fusion protein-attachment protein receptor (SNARE) protein-regulated exocytosis. SNARE complex assembly further requires the regulatory proteins Munc18c, Munc18-1 and Doc2b. Munc18-1 and Munc18c are required for first- and second-phase GSIS respectively. These distinct Munc18-1 and Munc18c roles are related to their transient high-affinity binding with their cognate target (t-)SNAREs, Syntaxin 1A and Syntaxin 4 respectively. Doc2b is essential for both phases of GSIS, yet the molecular basis for this remains unresolved. Because Doc2b binds to Munc18-1 and Munc18c via its distinct C2A and C2B domains respectively, we hypothesized that Doc2b may provide a plasma membrane-localized scaffold/platform for transient docking of these Munc18 isoforms during GSIS. Towards this, macromolecular complexes composed of Munc18c, Doc2b and Munc18-1 were detected in β-cells. In vitro interaction assays indicated that Doc2b is required to bridge the interaction between Munc18c and Munc18-1 in the macromolecular complex; Munc18c and Munc18-1 failed to associate in the absence of Doc2b. Competition-based GST-Doc2b interaction assays revealed that Doc2b could simultaneously bind both Munc18-1 and Munc18c. Hence these data support a working model wherein Doc2b functions as a docking platform/scaffold for transient interactions with the multiple Munc18 isoforms operative in insulin release, promoting SNARE assembly.
Collapse
|
47
|
MacDonald MJ, Ade L, Ntambi JM, Ansari IUH, Stoker SW. Characterization of phospholipids in insulin secretory granules and mitochondria in pancreatic beta cells and their changes with glucose stimulation. J Biol Chem 2015; 290:11075-92. [PMID: 25762724 DOI: 10.1074/jbc.m114.628420] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Indexed: 01/05/2023] Open
Abstract
The lipid composition of insulin secretory granules (ISG) has never previously been thoroughly characterized. We characterized the phospholipid composition of ISG and mitochondria in pancreatic beta cells without and with glucose stimulation. The phospholipid/protein ratios of most phospholipids containing unsaturated fatty acids were higher in ISG than in whole cells and in mitochondria. The concentrations of negatively charged phospholipids, phosphatidylserine, and phosphatidylinositol in ISG were 5-fold higher than in the whole cell. In ISG phosphatidylserine, phosphatidylinositol, phosphatidylethanolamine, and sphingomyelin, fatty acids 12:0 and 14:0 were high, as were phosphatidylserine and phosphatidylinositol containing 18-carbon unsaturated FA. With glucose stimulation, the concentration of many ISG phosphatidylserines and phosphatidylinositols increased; unsaturated fatty acids in phosphatidylserine increased; and most phosphatidylethanolamines, phosphatidylcholines, sphingomyelins, and lysophosphatidylcholines were unchanged. Unsaturation and shorter fatty acid length in phospholipids facilitate curvature and fluidity of membranes, which favors fusion of membranes. Recent evidence suggests that negatively charged phospholipids, such as phosphatidylserine, act as coupling factors enhancing the interaction of positively charged regions in SNARE proteins in synaptic or secretory vesicle membrane lipid bilayers with positively charged regions in SNARE proteins in the plasma membrane lipid bilayer to facilitate docking of vesicles to the plasma membrane during exocytosis. The results indicate that ISG phospholipids are in a dynamic state and are consistent with the idea that changes in ISG phospholipids facilitate fusion of ISG with the plasma membrane-enhancing glucose-stimulated insulin exocytosis.
Collapse
Affiliation(s)
- Michael J MacDonald
- From the Children's Diabetes Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53706 and
| | | | - James M Ntambi
- the Departments of Biochemistry and Nutritional Sciences, University of Wisconsin, Madison, Wisconsin 53706
| | - Israr-Ul H Ansari
- From the Children's Diabetes Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53706 and
| | - Scott W Stoker
- From the Children's Diabetes Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53706 and
| |
Collapse
|
48
|
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: 2.2] [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
|
49
|
Wuttke A. Lipid Signalling Dynamics at the β-cell Plasma Membrane. Basic Clin Pharmacol Toxicol 2015; 116:281-90. [DOI: 10.1111/bcpt.12369] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 12/15/2014] [Indexed: 12/26/2022]
Affiliation(s)
- Anne Wuttke
- Department of Medical Cell Biology; Uppsala University; Uppsala Sweden
| |
Collapse
|
50
|
Gaisano HY. Here come the newcomer granules, better late than never. Trends Endocrinol Metab 2014; 25:381-8. [PMID: 24746186 DOI: 10.1016/j.tem.2014.03.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 03/06/2014] [Accepted: 03/14/2014] [Indexed: 01/03/2023]
Abstract
Exocytosis in pancreatic β-cells employs Munc18/soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes that mediate the priming and docking onto the plasma membrane (PM) of insulin granules, called predocked granules, that sit on the PM until Ca(2+) influx evokes fusion. This accounts for most of the initial peak secretory response. However, the subsequent sustained phase of glucose-stimulated insulin secretion arises from newcomer granules that have a minimal residence time at the PM before fusion. In this Opinion I discuss recent work that has begun to decipher the components of the exocytotic machinery of newcomer granules, including a Munc18/SNARE complex that is different from that mediating the fusion of predocked granules and which can potentially rescue defective insulin secretion in diabetes. These insights are applicable to other neuroendocrine cells that exhibit sustained secretion.
Collapse
Affiliation(s)
- Herbert Y Gaisano
- Department of Medicine, University of Toronto, M5S 1A8, Toronto, Canada.
| |
Collapse
|