1
|
Sumsion JS, Shumway SW, Blocker TM, Weed TD, Chambers TM, Poland RJ, Woodbury DJ. Multiple structural states in an intrinsically disordered protein, SNAP-25, using circular dichroism. Biophys J 2025:S0006-3495(25)00060-8. [PMID: 39923128 DOI: 10.1016/j.bpj.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/22/2024] [Accepted: 02/05/2025] [Indexed: 02/10/2025] Open
Abstract
SNAP-25, together with other SNARE proteins, drives fusion of synaptic vesicles with the nerve cell membrane, leading to neurotransmitter release. It is unique in contributing two α helices to the four-helix bundle known as the SNARE complex. Complex formation drives fusion as these proteins transform from a disordered to ordered (coiled-coil) state. SNAP-25 has two isoforms, -25A and -25B, but little is known of any structural differences, nor are there extensive reports of the structures of its two helical domains, SN1 and SN2. Thus, the benefit of having two distinct isoforms of SNAP-25, each with two distinct domains, is unknown. Here, we use circular dichroism spectroscopy and mass spectrometry to further characterize the secondary structure of SNAP-25A, SNAP-25B, SN1, SN2, and a cysteine-free version of SNAP-25A. We demonstrate that these proteins undergo structural transitions, with changing fractions of α helix, β sheet, and random coil. These different structures can be induced by varying the environmental conditions of ionic strength, pH, temperature, or redox state. We use triangle plots to directly display the change in ternary composition following changes in these four parameters. We report that SNAP-25A and SNAP-25B make distinctly different structural changes. We show that the secondary structure of SN1 is more variable than SN2. These data add to the ongoing literature characterizing SNAP-25 as an intrinsically disordered protein that is sensitive to environmental conditions in neuronal cells and may function as a redox sensor to modulate neurotransmitter release.
Collapse
Affiliation(s)
- Jarom S Sumsion
- Department of Cell Biology & Physiology, Brigham Young University, Provo, Utah
| | - Samuel W Shumway
- Lake Erie College of Osteopathic Medicine, Greensburg, Pennsylvania
| | - Tanner M Blocker
- Department of Cell Biology & Physiology, Brigham Young University, Provo, Utah
| | - Thomas D Weed
- Department of Cell Biology & Physiology, Brigham Young University, Provo, Utah
| | | | - Ryan J Poland
- Neuroscience Center, Brigham Young University, Provo, Utah
| | - Dixon J Woodbury
- Department of Cell Biology & Physiology, Brigham Young University, Provo, Utah; Neuroscience Center, Brigham Young University, Provo, Utah.
| |
Collapse
|
2
|
LaForce GR, Philippidou P, Schaffer AE. mRNA isoform balance in neuronal development and disease. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023; 14:e1762. [PMID: 36123820 PMCID: PMC10024649 DOI: 10.1002/wrna.1762] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/11/2022] [Accepted: 08/15/2022] [Indexed: 11/07/2022]
Abstract
Balanced mRNA isoform diversity and abundance are spatially and temporally regulated throughout cellular differentiation. The proportion of expressed isoforms contributes to cell type specification and determines key properties of the differentiated cells. Neurons are unique cell types with intricate developmental programs, characteristic cellular morphologies, and electrophysiological potential. Neuron-specific gene expression programs establish these distinctive cellular characteristics and drive diversity among neuronal subtypes. Genes with neuron-specific alternative processing are enriched in key neuronal functions, including synaptic proteins, adhesion molecules, and scaffold proteins. Despite the similarity of neuronal gene expression programs, each neuronal subclass can be distinguished by unique alternative mRNA processing events. Alternative processing of developmentally important transcripts alters coding and regulatory information, including interaction domains, transcript stability, subcellular localization, and targeting by RNA binding proteins. Fine-tuning of mRNA processing is essential for neuronal activity and maintenance. Thus, the focus of neuronal RNA biology research is to dissect the transcriptomic mechanisms that underlie neuronal homeostasis, and consequently, predispose neuronal subtypes to disease. This article is categorized under: RNA in Disease and Development > RNA in Disease RNA in Disease and Development > RNA in Development.
Collapse
Affiliation(s)
- Geneva R LaForce
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Polyxeni Philippidou
- Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Ashleigh E Schaffer
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
3
|
Mertins J, Finke J, Sies R, Rink KM, Hasenauer J, Lang T. The mesoscale organization of syntaxin 1A and SNAP25 is determined by SNARE-SNARE interactions. eLife 2021; 10:69236. [PMID: 34779769 PMCID: PMC8629428 DOI: 10.7554/elife.69236] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 11/14/2021] [Indexed: 01/01/2023] Open
Abstract
SNARE proteins have been described as the effectors of fusion events in the secretory pathway more than two decades ago. The strong interactions between SNARE domains are clearly important in membrane fusion, but it is unclear whether they are involved in any other cellular processes. Here, we analyzed two classical SNARE proteins, syntaxin 1A and SNAP25. Although they are supposed to be engaged in tight complexes, we surprisingly find them largely segregated in the plasma membrane. Syntaxin 1A only occupies a small fraction of the plasma membrane area. Yet, we find it is able to redistribute the far more abundant SNAP25 on the mesoscale by gathering crowds of SNAP25 molecules onto syntaxin clusters in a SNARE-domain-dependent manner. Our data suggest that SNARE domain interactions are not only involved in driving membrane fusion on the nanoscale, but also play an important role in controlling the general organization of proteins on the mesoscale. Further, we propose these mechanisms preserve active syntaxin 1A–SNAP25 complexes at the plasma membrane.
Collapse
Affiliation(s)
- Jasmin Mertins
- Departments of Membrane Biochemistry, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Jérôme Finke
- Departments of Membrane Biochemistry, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Ricarda Sies
- Departments of Membrane Biochemistry, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Kerstin M Rink
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany
| | - Jan Hasenauer
- Computational Life Sciences, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.,Interdisciplinary Research Unit Mathematics and Life Sciences, University of Bonn, Bonn, Germany.,Institute of Computational Biology, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Germany
| | - Thorsten Lang
- Departments of Membrane Biochemistry, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
4
|
Dudek KD, Osipovich AB, Cartailler JP, Gu G, Magnuson MA. Insm1, Neurod1, and Pax6 promote murine pancreatic endocrine cell development through overlapping yet distinct RNA transcription and splicing programs. G3-GENES GENOMES GENETICS 2021; 11:6358139. [PMID: 34534285 PMCID: PMC8527475 DOI: 10.1093/g3journal/jkab303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/19/2021] [Indexed: 11/13/2022]
Abstract
Insm1, Neurod1, and Pax6 are essential for the formation and function of pancreatic endocrine cells. Here, we report comparative immunohistochemical, transcriptomic, functional enrichment, and RNA splicing analyses of these genes using gene knock-out mice. Quantitative immunohistochemical analysis confirmed that elimination of each of these three factors variably impairs the proliferation, survival, and differentiation of endocrine cells. Transcriptomic analysis revealed that each factor contributes uniquely to the transcriptome although their effects were overlapping. Functional enrichment analysis revealed that genes downregulated by the elimination of Insm1, Neurod1, and Pax6 are commonly involved in mRNA metabolism, chromatin organization, secretion, and cell cycle regulation, and upregulated genes are associated with protein degradation, autophagy, and apoptotic process. Elimination of Insm1, Neurod1, and Pax6 impaired expression of many RNA-binding proteins thereby altering RNA splicing events, including for Syt14 and Snap25, two genes required for insulin secretion. All three factors are necessary for normal splicing of Syt14, and both Insm1 and Pax6 are necessary for the processing of Snap25. Collectively, these data provide new insights into how Insm1, Neurod1, and Pax6 contribute to the formation of functional pancreatic endocrine cells.
Collapse
Affiliation(s)
- Karrie D Dudek
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA.,Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Anna B Osipovich
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | | | - Guoquing Gu
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA.,Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Mark A Magnuson
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA.,Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
5
|
Milosevic I. Spatial and Temporal Aspects of Exocytosis Studied on the Isolated Plasma Membranes. Methods Mol Biol 2021; 2233:311-325. [PMID: 33222144 DOI: 10.1007/978-1-0716-1044-2_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Exocytosis of large-dense core vesicles in neuroendocrine cells is a highly regulated, calcium-dependent process, mediated by networks of interrelated proteins and lipids. Here, I describe experimental procedures for studies of selective spatial and temporal aspects of exocytosis at the plasma membrane, or in its proximity, using adrenal chromaffin cells. The assay utilizes primary cells subjected to a brief ultrasonic pulse, resulting in the formation of thin, flat inside-out plasma membranes with attached secretory vesicles and elements of cell cytoskeleton. In this model, secretion of plasma membrane-attached secretory vesicles was found to be dependent on calcium and sensitive to clostridial neurotoxins. Depending on the probe selected for secretory vesicle cargo, protein, and/or lipid detection, this simple assay is versatile, fast and inexpensive, and offers excellent spatial resolution.
Collapse
Affiliation(s)
- Ira Milosevic
- European Neuroscience Institute (ENI), A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society, Göttingen, Germany. .,Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK.
| |
Collapse
|
6
|
McGrowder DA, Miller F, Vaz K, Nwokocha C, Wilson-Clarke C, Anderson-Cross M, Brown J, Anderson-Jackson L, Williams L, Latore L, Thompson R, Alexander-Lindo R. Cerebrospinal Fluid Biomarkers of Alzheimer's Disease: Current Evidence and Future Perspectives. Brain Sci 2021; 11:215. [PMID: 33578866 PMCID: PMC7916561 DOI: 10.3390/brainsci11020215] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease is a progressive, clinically heterogeneous, and particularly complex neurodegenerative disease characterized by a decline in cognition. Over the last two decades, there has been significant growth in the investigation of cerebrospinal fluid (CSF) biomarkers for Alzheimer's disease. This review presents current evidence from many clinical neurochemical studies, with findings that attest to the efficacy of existing core CSF biomarkers such as total tau, phosphorylated tau, and amyloid-β (Aβ42), which diagnose Alzheimer's disease in the early and dementia stages of the disorder. The heterogeneity of the pathophysiology of the late-onset disease warrants the growth of the Alzheimer's disease CSF biomarker toolbox; more biomarkers showing other aspects of the disease mechanism are needed. This review focuses on new biomarkers that track Alzheimer's disease pathology, such as those that assess neuronal injury (VILIP-1 and neurofilament light), neuroinflammation (sTREM2, YKL-40, osteopontin, GFAP, progranulin, and MCP-1), synaptic dysfunction (SNAP-25 and GAP-43), vascular dysregulation (hFABP), as well as CSF α-synuclein levels and TDP-43 pathology. Some of these biomarkers are promising candidates as they are specific and predict future rates of cognitive decline. Findings from the combinations of subclasses of new Alzheimer's disease biomarkers that improve their diagnostic efficacy in detecting associated pathological changes are also presented.
Collapse
Affiliation(s)
- Donovan A. McGrowder
- Department of Pathology, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (K.V.); (J.B.); (L.A.-J.); (L.L.); (R.T.)
| | - Fabian Miller
- Department of Physical Education, Faculty of Education, The Mico University College, 1A Marescaux Road, Kingston 5, Jamaica;
- Department of Biotechnology, Faculty of Science and Technology, The University of the West Indies, Kingston 7, Jamaica;
| | - Kurt Vaz
- Department of Pathology, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (K.V.); (J.B.); (L.A.-J.); (L.L.); (R.T.)
| | - Chukwuemeka Nwokocha
- Department of Basic Medical Sciences, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (C.N.); (C.W.-C.); (R.A.-L.)
| | - Cameil Wilson-Clarke
- Department of Basic Medical Sciences, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (C.N.); (C.W.-C.); (R.A.-L.)
| | - Melisa Anderson-Cross
- School of Allied Health and Wellness, College of Health Sciences, University of Technology, Kingston 7, Jamaica;
| | - Jabari Brown
- Department of Pathology, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (K.V.); (J.B.); (L.A.-J.); (L.L.); (R.T.)
| | - Lennox Anderson-Jackson
- Department of Pathology, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (K.V.); (J.B.); (L.A.-J.); (L.L.); (R.T.)
| | - Lowen Williams
- Department of Biotechnology, Faculty of Science and Technology, The University of the West Indies, Kingston 7, Jamaica;
| | - Lyndon Latore
- Department of Pathology, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (K.V.); (J.B.); (L.A.-J.); (L.L.); (R.T.)
| | - Rory Thompson
- Department of Pathology, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (K.V.); (J.B.); (L.A.-J.); (L.L.); (R.T.)
| | - Ruby Alexander-Lindo
- Department of Basic Medical Sciences, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica; (C.N.); (C.W.-C.); (R.A.-L.)
| |
Collapse
|
7
|
Gowrisankaran S, Houy S, Del Castillo JGP, Steubler V, Gelker M, Kroll J, Pinheiro PS, Schwitters D, Halbsgut N, Pechstein A, van Weering JRT, Maritzen T, Haucke V, Raimundo N, Sørensen JB, Milosevic I. Endophilin-A coordinates priming and fusion of neurosecretory vesicles via intersectin. Nat Commun 2020; 11:1266. [PMID: 32152276 PMCID: PMC7062783 DOI: 10.1038/s41467-020-14993-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 02/12/2020] [Indexed: 12/17/2022] Open
Abstract
Endophilins-A are conserved endocytic adaptors with membrane curvature-sensing and -inducing properties. We show here that, independently of their role in endocytosis, endophilin-A1 and endophilin-A2 regulate exocytosis of neurosecretory vesicles. The number and distribution of neurosecretory vesicles were not changed in chromaffin cells lacking endophilin-A, yet fast capacitance and amperometry measurements revealed reduced exocytosis, smaller vesicle pools and altered fusion kinetics. The levels and distributions of the main exocytic and endocytic factors were unchanged, and slow compensatory endocytosis was not robustly affected. Endophilin-A’s role in exocytosis is mediated through its SH3-domain, specifically via a direct interaction with intersectin-1, a coordinator of exocytic and endocytic traffic. Endophilin-A not able to bind intersectin-1, and intersectin-1 not able to bind endophilin-A, resulted in similar exocytic defects in chromaffin cells. Altogether, we report that two endocytic proteins, endophilin-A and intersectin-1, are enriched on neurosecretory vesicles and regulate exocytosis by coordinating neurosecretory vesicle priming and fusion. Endophilins-A are conserved membrane-associated proteins required for endocytosis. Here, the authors report that endophilins-A also promote exocytosis of neurosecretory vesicles by coordinating priming and fusion through intersectin-1, independently of their roles in different types of endocytosis.
Collapse
Affiliation(s)
- Sindhuja Gowrisankaran
- European Neuroscience Institute-A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society Göttingen, Göttingen, Germany
| | - Sébastien Houy
- University of Copenhagen, Department for Neuroscience, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Johanna G Peña Del Castillo
- European Neuroscience Institute-A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society Göttingen, Göttingen, Germany
| | - Vicky Steubler
- European Neuroscience Institute-A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society Göttingen, Göttingen, Germany
| | - Monika Gelker
- European Neuroscience Institute-A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society Göttingen, Göttingen, Germany
| | - Jana Kroll
- European Neuroscience Institute-A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society Göttingen, Göttingen, Germany
| | - Paulo S Pinheiro
- University of Copenhagen, Department for Neuroscience, Faculty of Health and Medical Sciences, Copenhagen, Denmark.,Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Dirk Schwitters
- European Neuroscience Institute-A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society Göttingen, Göttingen, Germany
| | - Nils Halbsgut
- European Neuroscience Institute-A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society Göttingen, Göttingen, Germany
| | - Arndt Pechstein
- Leibniz Research Institute for Molecular Pharmacology, Molecular Physiology and Cell Biology Section, Berlin, Germany
| | - Jan R T van Weering
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam UMC, Amsterdam, The Netherlands
| | - Tanja Maritzen
- Leibniz Research Institute for Molecular Pharmacology, Molecular Physiology and Cell Biology Section, Berlin, Germany
| | - Volker Haucke
- Leibniz Research Institute for Molecular Pharmacology, Molecular Physiology and Cell Biology Section, Berlin, Germany
| | - Nuno Raimundo
- Institute for Cellular Biochemistry, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Jakob B Sørensen
- University of Copenhagen, Department for Neuroscience, Faculty of Health and Medical Sciences, Copenhagen, Denmark.
| | - Ira Milosevic
- European Neuroscience Institute-A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society Göttingen, Göttingen, Germany.
| |
Collapse
|
8
|
Gopaul KR, Irfan M, Miry O, Vose LR, Moghadam A, Subah G, Hökfelt T, Bark C, Stanton PK. Developmental Time Course of SNAP-25 Isoforms Regulate Hippocampal Long-Term Synaptic Plasticity and Hippocampus-Dependent Learning. Int J Mol Sci 2020; 21:ijms21041448. [PMID: 32093363 PMCID: PMC7073020 DOI: 10.3390/ijms21041448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/12/2020] [Accepted: 02/18/2020] [Indexed: 12/28/2022] Open
Abstract
SNAP-25 is essential to activity-dependent vesicle fusion and neurotransmitter release in the nervous system. During early development and adulthood, SNAP-25 appears to have differential influences on short- and long-term synaptic plasticity. The involvement of SNAP-25 in these processes may be different at hippocampal and neocortical synapses because of the presence of two different splice variants, which are developmentally regulated. We show here that the isoform SNAP-25a, which is expressed first developmentally in rodent brain, contributes to developmental regulation of the expression of both long-term depression (LTD) and long-term potentiation (LTP) at Schaffer collateral-CA1 synapses in the hippocampus. In one month old mice lacking the developmentally later expressed isoform SNAP-25b, Schaffer collateral-CA1 synapses showed faster release kinetics, decreased LTP and enhanced LTD. By four months of age, SNAP-25b-deficient mice appeared to have compensated for the lack of the adult SNAP-25b isoform, now exhibiting larger LTP and no differences in LTD compared to wild type mice. Interestingly, learning a hippocampus-dependent task reversed the reductions in LTP, but not LTD, seen at one month of age. In four month old adult mice, learning prevented the compensatory up-regulation of LTD that we observed prior to training. These findings support the hypothesis that SNAP-25b promotes stronger LTP and weakens LTD at Schaffer collateral-CA1 synapses in young mice, and suggest that compensatory mechanisms can reverse alterations in synaptic plasticity associated with a lack of SNAP-25b, once mice reach adulthood.
Collapse
Affiliation(s)
- Katisha R. Gopaul
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY 10595, USA; (K.R.G.); (M.I.); (O.M.); (L.R.V.); (A.M.); (G.S.)
| | - Muhammad Irfan
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY 10595, USA; (K.R.G.); (M.I.); (O.M.); (L.R.V.); (A.M.); (G.S.)
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden;
| | - Omid Miry
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY 10595, USA; (K.R.G.); (M.I.); (O.M.); (L.R.V.); (A.M.); (G.S.)
| | - Linnea R. Vose
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY 10595, USA; (K.R.G.); (M.I.); (O.M.); (L.R.V.); (A.M.); (G.S.)
| | - Alexander Moghadam
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY 10595, USA; (K.R.G.); (M.I.); (O.M.); (L.R.V.); (A.M.); (G.S.)
| | - Galadu Subah
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY 10595, USA; (K.R.G.); (M.I.); (O.M.); (L.R.V.); (A.M.); (G.S.)
| | - Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden;
| | - Christina Bark
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden;
- Correspondence: (C.B.); (P.K.S.); Tel. +46-085-248-6984 (C.B.); +1-914-594-4883 (P.K.S.)
| | - Patric K. Stanton
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY 10595, USA; (K.R.G.); (M.I.); (O.M.); (L.R.V.); (A.M.); (G.S.)
- Correspondence: (C.B.); (P.K.S.); Tel. +46-085-248-6984 (C.B.); +1-914-594-4883 (P.K.S.)
| |
Collapse
|
9
|
Ruiter M, Houy S, Engholm-Keller K, Graham ME, Sørensen JB. SNAP-25 phosphorylation at Ser187 is not involved in Ca 2+ or phorbolester-dependent potentiation of synaptic release. Mol Cell Neurosci 2019; 102:103452. [PMID: 31794878 DOI: 10.1016/j.mcn.2019.103452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 10/13/2019] [Accepted: 11/29/2019] [Indexed: 11/25/2022] Open
Abstract
SNAP-25, one of the three SNARE-proteins responsible for synaptic release, can be phosphorylated by Protein Kinase C on Ser-187, close to the fusion pore. In neuroendocrine cells, this phosphorylation event potentiates vesicle recruitment into releasable pools, whereas the consequences of phosphorylation for synaptic release remain unclear. We mutated Ser-187 and expressed two mutants (S187C and S187E) in the context of the SNAP-25B-isoform in SNAP-25 knockout glutamatergic autaptic neurons. Whole-cell patch clamp recordings were performed to assess the effect of Ser-187 phosphorylation on synaptic transmission. Blocking phosphorylation by expressing the S187C mutant did not affect synapse density, basic evoked or spontaneous neurotransmission, the readily-releasable pool size or its Ca2+-independent or Ca2+-dependent replenishment. Furthermore, it did not affect the response to phorbol esters, which activate PKC. Expressing S187C in the context of the SNAP-25A isoform also did not affect synaptic transmission. Strikingly, the - potentially phosphomimetic - mutant S187E reduced spontaneous release and release probability, with the largest effect seen in the SNAP-25B isoform, showing that a negative charge in this position is detrimental for neurotransmission, in agreement with electrostatic fusion triggering. During the course of our experiments, we found that higher SNAP-25B expression levels led to decreased paired pulse potentiation, probably due to higher release probabilities. Under these conditions, the potentiation of evoked EPSCs by phorbol esters was followed by a persistent down-regulation, probably due to a ceiling effect. In conclusion, our results indicate that phosphorylation of Ser-187 in SNAP-25 is not involved in modulation of synaptic release by Ca2+ or phorbol esters.
Collapse
Affiliation(s)
- Marvin Ruiter
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3C, 2200 Copenhagen N, Denmark
| | - Sébastien Houy
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3C, 2200 Copenhagen N, Denmark
| | - Kasper Engholm-Keller
- Synapse Proteomics Group, Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead NSW 2145, New South Wales, Australia; Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Mark E Graham
- Synapse Proteomics Group, Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead NSW 2145, New South Wales, Australia
| | - Jakob B Sørensen
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3C, 2200 Copenhagen N, Denmark.
| |
Collapse
|
10
|
The SNAP-25 Protein Family. Neuroscience 2019; 420:50-71. [DOI: 10.1016/j.neuroscience.2018.09.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/31/2018] [Accepted: 09/14/2018] [Indexed: 01/04/2023]
|
11
|
Kroeze Y, Oti M, van Beusekom E, Cooijmans RHM, van Bokhoven H, Kolk SM, Homberg JR, Zhou H. Transcriptome Analysis Identifies Multifaceted Regulatory Mechanisms Dictating a Genetic Switch from Neuronal Network Establishment to Maintenance During Postnatal Prefrontal Cortex Development. Cereb Cortex 2019; 28:833-851. [PMID: 28108491 DOI: 10.1093/cercor/bhw407] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Indexed: 12/20/2022] Open
Abstract
The prefrontal cortex (PFC) is one of the latest brain regions to mature, which allows the acquisition of complex cognitive abilities through experience. To unravel the underlying gene expression changes during postnatal development, we performed RNA-sequencing (RNA-seq) in the rat medial PFC (mPFC) at five developmental time points from infancy to adulthood, and analyzed the differential expression of protein-coding genes, long intergenic noncoding RNAs (lincRNAs), and alternative exons. We showed that most expression changes occur in infancy, and that the number of differentially expressed genes reduces toward adulthood. We observed 137 differentially expressed lincRNAs and 796 genes showing alternative exon usage during postnatal development. Importantly, we detected a genetic switch from neuronal network establishment in infancy to maintenance of neural networks in adulthood based on gene expression dynamics, involving changes in protein-coding and lincRNA gene expression as well as alternative exon usage. Our gene expression datasets provide insights into the multifaceted transcriptional regulation of the developing PFC. They can be used to study the basic developmental processes of the mPFC and to understand the mechanisms of neurodevelopmental and neuropsychiatric disorders. Our study provides an important contribution to the ongoing efforts to complete the "brain map", and to the understanding of PFC development.
Collapse
Affiliation(s)
- Yvet Kroeze
- Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6525 EZ Nijmegen, The Netherlands.,Department of Human Genetics, Centre for Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands
| | - Martin Oti
- Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, 6525 GA Nijmegen, The Netherlands.,Carlos Chagas Filho Biophysics Institute (IBCCF), Federal University of Rio de Janeiro (UFRJ), 21941-902 Rio de Janeiro, Brazil
| | - Ellen van Beusekom
- Department of Human Genetics, Centre for Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands
| | - Roel H M Cooijmans
- Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, 6525 GA Nijmegen, The Netherlands
| | - Hans van Bokhoven
- Department of Human Genetics, Centre for Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands
| | - Sharon M Kolk
- Department of Molecular Animal Physiology, Centre for Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University, 6525 GA Nijmegen, The Netherlands
| | - Judith R Homberg
- Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6525 EZ Nijmegen, The Netherlands
| | - Huiqing Zhou
- Department of Human Genetics, Centre for Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands.,Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
12
|
Daraio T, Valladolid-Acebes I, Brismar K, Bark C. SNAP-25a and SNAP-25b differently mediate interactions with Munc18-1 and Gβγ subunits. Neurosci Lett 2018; 674:75-80. [PMID: 29548989 DOI: 10.1016/j.neulet.2018.03.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 03/09/2018] [Accepted: 03/12/2018] [Indexed: 02/06/2023]
Abstract
SNAP-25 is a protein involved in regulated membrane fusion and part of the SNARE complex. It exists as two splicing variants, SNAP-25a and SNAP-25b, which differ in 9 out of 206 amino acids. SNAP-25 together with Syntaxin 1 and VAMP-2 forms the ternary SNARE complex essential for mediating activity-dependent release of hormones and neurotransmitters. The functional difference between SNAP-25a and SNAP-25b is poorly understood as both can participate in SNARE complexes and mediate membrane fusion. However, we recently demonstrated that SNAP-25b-deficiency results in metabolic disease and increased insulin secretion. Here we investigated if SNAP-25a and SNAP-25b differently affect interactions with other SNAREs and SNARE-interacting proteins in mouse hippocampus. Adult mice almost exclusively express the SNAP-25b protein in hippocampus whereas SNAP-25b-deficient mice only express SNAP-25a. Immunoprecipitation studies showed no significant differences in amount of Syntaxin 1 and VAMP-2 co-precipitated with the different SNAP-25 isoforms. In contrast, Munc18-1, that preferentially interacts with SNAP-25 via Syntaxin 1 and/or the trimeric SNARE complex, demonstrated an increased ability to bind protein-complexes containing SNAP-25b. Moreover, we found that both SNAP-25 isoforms co-precipitated the Gβγ subunits of the heterotrimeric G proteins, an interaction known to play a role in presynaptic inhibition. We have identified Gβ1 and Gβ2 as the interacting partners of both SNAP-25 isoforms in mouse hippocampus, but Gβ2 was less efficiently captured by SNAP-25a. These results implicate that the two SNAP-25 isoforms could differently mediate protein interactions outside the ternary SNARE core complex and thereby contribute to modulate neurotransmission.
Collapse
Affiliation(s)
- Teresa Daraio
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Ismael Valladolid-Acebes
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Kerstin Brismar
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Christina Bark
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE-171 76 Stockholm, Sweden.
| |
Collapse
|
13
|
Spatial and Temporal Aspects of Phosphoinositides in Endocytosis Studied in the Isolated Plasma Membranes. Methods Mol Biol 2018; 1847:147-160. [PMID: 30129015 DOI: 10.1007/978-1-4939-8719-1_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Endocytosis is a well-orchestrated cascade of lipid-protein and protein-protein interactions resulting in formation and internalization of vesicles. Membrane phospholipids have key regulatory functions in endocytosis and membrane traffic. I have previously described an in vitro assay based on the isolated, substrate-attached plasma membrane to study the spatial distribution and levels of phosphoinositides, in particular phosphatidylinositol-4,5-bisphospate [PI(4,5)P2]. This assay utilizes cultured cells subjected to a brief ultrasonic pulse, resulting in the formation of thin, flat inside-out plasma membrane sheets with elements of cell cytoskeleton. Here, I describe an experimental procedure for "on-stage" and "off-stage" detection of PI(4,5)P2 spatial distribution, and semi-quantification of PI(4,5)P2 levels in the plasma membrane using fluorescence microscopy. Depending on the probe selected for lipid detection, this simple assay can be modified to study other plasmalemmal phospholipids and/or proteins.
Collapse
|
14
|
Yang Z, Gou L, Chen S, Li N, Zhang S, Zhang L. Membrane Fusion Involved in Neurotransmission: Glimpse from Electron Microscope and Molecular Simulation. Front Mol Neurosci 2017. [PMID: 28638320 PMCID: PMC5461332 DOI: 10.3389/fnmol.2017.00168] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Membrane fusion is one of the most fundamental physiological processes in eukaryotes for triggering the fusion of lipid and content, as well as the neurotransmission. However, the architecture features of neurotransmitter release machinery and interdependent mechanism of synaptic membrane fusion have not been extensively studied. This review article expounds the neuronal membrane fusion processes, discusses the fundamental steps in all fusion reactions (membrane aggregation, membrane association, lipid rearrangement and lipid and content mixing) and the probable mechanism coupling to the delivery of neurotransmitters. Subsequently, this work summarizes the research on the fusion process in synaptic transmission, using electron microscopy (EM) and molecular simulation approaches. Finally, we propose the future outlook for more exciting applications of membrane fusion involved in synaptic transmission, with the aid of stochastic optical reconstruction microscopy (STORM), cryo-EM (cryo-EM), and molecular simulations.
Collapse
Affiliation(s)
- Zhiwei Yang
- Department of Applied Physics, Xi'an Jiaotong UniversityXi'an, China.,Department of Applied Chemistry, Xi'an Jiaotong UniversityXi'an, China.,School of Life Science and Technology, Xi'an Jiaotong UniversityXi'an, China
| | - Lu Gou
- Department of Applied Physics, Xi'an Jiaotong UniversityXi'an, China
| | - Shuyu Chen
- Department of Applied Physics, Xi'an Jiaotong UniversityXi'an, China
| | - Na Li
- Department of Applied Physics, Xi'an Jiaotong UniversityXi'an, China
| | - Shengli Zhang
- Department of Applied Physics, Xi'an Jiaotong UniversityXi'an, China
| | - Lei Zhang
- Department of Applied Physics, Xi'an Jiaotong UniversityXi'an, China
| |
Collapse
|
15
|
Neuronal activity-regulated alternative mRNA splicing. Int J Biochem Cell Biol 2017; 91:184-193. [PMID: 28591617 DOI: 10.1016/j.biocel.2017.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/24/2017] [Accepted: 06/01/2017] [Indexed: 11/20/2022]
Abstract
Activity-regulated gene transcription underlies plasticity-dependent changes in the molecular composition and structure of neurons. Numerous genes whose expression is induced by different neuronal plasticity inducing pathways have been identified, but the alteration of gene expression levels represents only part of the complexity of the activity-regulated transcriptional program. Alternative splicing of precursor mRNA is an additional mechanism that modulates the activity-dependent transcriptional signature. Recently developed splicing sensitive transcriptome wide analyses improve our understanding of the underlying mechanisms and demonstrate to what extend the activity regulated transcriptome is alternatively spliced. So far, only for a small group of differentially spliced mRNAs of synaptic proteins, the functional implications have been studied in detail. These include examples in which differential exon usage can result in the expression of alternative proteins which interfere with or alter the function of preexisting proteins and cause a dominant negative functional block of constitutively expressed variants. Such altered proteins contribute to the structural and functional reorganization of pre- and postsynaptic terminals and to the maintenance and formation of synapses. In addition, activity-induced alternative splicing can affect the untranslated regions (UTRs) and generates mRNAs harboring different cis-regulatory elements. Such differential UTRs can influence mRNA stability, translation, and can change the targeting of mRNAs to subcellular compartments. Here, we summarize different categories of alternative splicing which are thought to contribute to synaptic remodeling, give an overview of activity-regulated alternatively spliced mRNAs of synaptic proteins that impact synaptic functions, and discuss splicing factors and epigenetic modifications as regulatory determinants.
Collapse
|
16
|
Uzuntarla M, Torres JJ, So P, Ozer M, Barreto E. Double inverse stochastic resonance with dynamic synapses. Phys Rev E 2017; 95:012404. [PMID: 28208458 DOI: 10.1103/physreve.95.012404] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Indexed: 06/06/2023]
Abstract
We investigate the behavior of a model neuron that receives a biophysically realistic noisy postsynaptic current based on uncorrelated spiking activity from a large number of afferents. We show that, with static synapses, such noise can give rise to inverse stochastic resonance (ISR) as a function of the presynaptic firing rate. We compare this to the case with dynamic synapses that feature short-term synaptic plasticity and show that the interval of presynaptic firing rate over which ISR exists can be extended or diminished. We consider both short-term depression and facilitation. Interestingly, we find that a double inverse stochastic resonance (DISR), with two distinct wells centered at different presynaptic firing rates, can appear.
Collapse
Affiliation(s)
- Muhammet Uzuntarla
- Department of Biomedical Engineering, Bulent Ecevit University, 67100 Zonguldak, Turkey
| | - Joaquin J Torres
- Department of Electromagnetism and Physics of the Matter and Institute Carlos I for Theoretical and Computational Physics, University of Granada, E-18071 Granada, Spain
| | - Paul So
- Department of Physics and Astronomy and the Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia 22030, USA
| | - Mahmut Ozer
- Department of Electrical and Electronics Engineering, Bulent Ecevit University, 67100 Zonguldak, Turkey
| | - Ernest Barreto
- Department of Physics and Astronomy and the Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia 22030, USA
| |
Collapse
|
17
|
Barakauskas VE, Moradian A, Barr AM, Beasley CL, Rosoklija G, Mann JJ, Ilievski B, Stankov A, Dwork AJ, Falkai P, Morin GB, Honer WG. Quantitative mass spectrometry reveals changes in SNAP-25 isoforms in schizophrenia. Schizophr Res 2016; 177:44-51. [PMID: 26971072 PMCID: PMC5017887 DOI: 10.1016/j.schres.2016.03.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/02/2016] [Accepted: 03/04/2016] [Indexed: 11/24/2022]
Abstract
SNAP-25 and syntaxin are presynaptic terminal SNARE proteins altered in amount and function in schizophrenia. In the ventral caudate, we observed 32% lower SNAP-25 and 26% lower syntaxin, but greater interaction between the two proteins using an in vitro assay. SNAP-25 has two isoforms, SNAP-25A and B, differing by only 9 amino acids, but with different effects on neurotransmission. A quantitative mass spectrometry assay was developed to measure total SNAP-25, and proportions of SNAP-25A and B. The assay had a good linear range (50- to 150-fold) and coefficient of variation (4.5%). We studied ventral caudate samples from patients with schizophrenia (n=15) previously reported to have lower total SNAP-25 than controls (n=13). We confirmed 27% lower total SNAP-25 in schizophrenia, and observed 31% lower SNAP-25A (P=0.002) with 20% lower SNAP-25B amounts (P=0.10). Lower SNAP-25A amount correlated with greater SNAP-25-syntaxin protein-protein interactions (r=-0.41, P=0.03); the level of SNAP-25B did not. Administration of haloperidol or clozapine to rats did not mimic the changes found in schizophrenia. The findings suggest that lower levels of SNAP-25 in schizophrenia may represent a greater effect of the illness on the SNAP-25A isoform. This in turn could contribute to the greater interaction between SNAP25 and syntaxin, and possibly disturb neurotransmission in the illness.
Collapse
Affiliation(s)
- Vilte E Barakauskas
- BC Mental Health and Addictions Research Institute, Vancouver, BC,Department of Psychiatry, University of British Columbia, Vancouver, BC
| | - Annie Moradian
- Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC
| | - Alasdair M. Barr
- BC Mental Health and Addictions Research Institute, Vancouver, BC,Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia, Vancouver, BC
| | - Clare L Beasley
- BC Mental Health and Addictions Research Institute, Vancouver, BC,Department of Psychiatry, University of British Columbia, Vancouver, BC
| | - Gorazd Rosoklija
- Department of Molecular Imaging and Neuropathology, New York State Psychiatric Institute; and Department of Psychiatry, Columbia University, New York, NY, USA,Macedonian Academy of Sciences and Arts, University “SS. Cyril and Methodius,” Skopje, Macedonia
| | - J John Mann
- Department of Molecular Imaging and Neuropathology, New York State Psychiatric Institute; and Department of Psychiatry, Columbia University, New York, NY, USA
| | - Boro Ilievski
- Department of Molecular Imaging and Neuropathology, New York State Psychiatric Institute; and Department of Psychiatry, Columbia University, New York, NY, USA,Institute for Pathology, University “SS. Cyril and Methodius,” Skopje, Macedonia
| | - Aleksandar Stankov
- Institute of Forensic Medicine, Criminology and Medical Deontology, University “SS. Cyril and Methodius,” Skopje, Macedonia
| | - Andrew J Dwork
- Department of Molecular Imaging and Neuropathology, New York State Psychiatric Institute; and Department of Psychiatry, Columbia University, New York, NY, USA,Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University, Munich, Germany
| | - Gregg B Morin
- Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.
| | - William G Honer
- BC Mental Health and Addictions Research Institute, Vancouver, BC, Canada; Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
18
|
Toft-Bertelsen TL, Ziomkiewicz I, Houy S, Pinheiro PS, Sørensen JB. Regulation of Ca2+ channels by SNAP-25 via recruitment of syntaxin-1 from plasma membrane clusters. Mol Biol Cell 2016; 27:3329-3341. [PMID: 27605709 PMCID: PMC5170865 DOI: 10.1091/mbc.e16-03-0184] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 09/01/2016] [Indexed: 12/20/2022] Open
Abstract
SNAP-25 regulates Ca2+ channels in an unknown manner. Endogenous and exogenous SNAP-25 inhibit Ca2+ currents indirectly by recruiting syntaxin-1 from clusters on the plasma membrane, thereby making it available for Ca2+ current inhibition. Thus the cell can regulate Ca2+ influx by expanding or contracting syntaxin-1 clusters. SNAP-25 regulates Ca2+ channels, with potentially important consequences for diseases involving an aberrant SNAP-25 expression level. How this regulation is executed mechanistically remains unknown. We investigated this question in mouse adrenal chromaffin cells and found that SNAP-25 inhibits Ca2+ currents, with the B-isoform being more potent than the A-isoform, but not when syntaxin-1 is cleaved by botulinum neurotoxin C. In contrast, syntaxin-1 inhibits Ca2+ currents independently of SNAP-25. Further experiments using immunostaining showed that endogenous or exogenous SNAP-25 expression recruits syntaxin-1 from clusters on the plasma membrane, thereby increasing the immunoavailability of syntaxin-1 and leading indirectly to Ca2+ current inhibition. Expression of Munc18-1, which recruits syntaxin-1 within the exocytotic pathway, does not modulate Ca2+ channels, whereas overexpression of the syntaxin-binding protein Doc2B or ubMunc13-2 increases syntaxin-1 immunoavailability and concomitantly down-regulates Ca2+ currents. Similar findings were obtained upon chemical cholesterol depletion, leading directly to syntaxin-1 cluster dispersal and Ca2+ current inhibition. We conclude that clustering of syntaxin-1 allows the cell to maintain a high syntaxin-1 expression level without compromising Ca2+ influx, and recruitment of syntaxin-1 from clusters by SNAP-25 expression makes it available for regulating Ca2+ channels. This mechanism potentially allows the cell to regulate Ca2+ influx by expanding or contracting syntaxin-1 clusters.
Collapse
Affiliation(s)
- Trine Lisberg Toft-Bertelsen
- Neurosecretion Group, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Iwona Ziomkiewicz
- Neurosecretion Group, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Sébastien Houy
- Neurosecretion Group, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Paulo S Pinheiro
- Neurosecretion Group, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Jakob B Sørensen
- Neurosecretion Group, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| |
Collapse
|
19
|
Molecular Features Underlying Neurodegeneration Identified through In Vitro Modeling of Genetically Diverse Parkinson's Disease Patients. Cell Rep 2016; 15:2411-26. [PMID: 27264186 DOI: 10.1016/j.celrep.2016.05.022] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 02/22/2016] [Accepted: 05/03/2016] [Indexed: 12/20/2022] Open
Abstract
The fact that Parkinson's disease (PD) can arise from numerous genetic mutations suggests a unifying molecular pathology underlying the various genetic backgrounds. To address this hypothesis, we took an integrated approach utilizing in vitro disease modeling and comprehensive transcriptome profiling to advance our understanding of PD progression and the concordant downstream signaling pathways across divergent genetic predispositions. To model PD in vitro, we generated neurons harboring disease-causing mutations from patient-specific, induced pluripotent stem cells (iPSCs). We observed signs of degeneration in midbrain dopaminergic neurons, reflecting the cardinal feature of PD. Gene expression signatures of PD neurons provided molecular insights into disease phenotypes observed in vitro, including oxidative stress vulnerability and altered neuronal activity. Notably, PD neurons show that elevated RBFOX1, a gene previously linked to neurodevelopmental diseases, underlies a pattern of alternative RNA-processing associated with PD-specific phenotypes.
Collapse
|
20
|
Liu YS, Dai X, Wu W, Yuan FF, Gu X, Chen JG, Zhu LQ, Wu J. The Association of SNAP25 Gene Polymorphisms in Attention Deficit/Hyperactivity Disorder: a Systematic Review and Meta-Analysis. Mol Neurobiol 2016; 54:2189-2200. [PMID: 26941099 DOI: 10.1007/s12035-016-9810-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 02/22/2016] [Indexed: 11/30/2022]
Abstract
Attention deficit/hyperactivity disorder (ADHD) is one of the most highly heritable psychiatric disorders in childhood. The risk gene mutation accounts for about 60 to 90 % cases. Synaptosomal-associated protein of 25 kDa (SNAP-25) is a presynaptic plasma membrane protein which is expressed highly and specifically in the neuronal cells. A number of evidences have suggested the role of SNAP-25 in the etiology of ADHD. Notably, the animal model of coloboma mouse mutant bears a ∼2-cM deletion encompassing genes including SNAP25 and displays spontaneous hyperkinetic behavior. Previous investigators have reported association between SNPs in SNAP25 and ADHD, and controversial results were observed. In this study, we analyzed the possible association between six polymorphisms (rs3746544, rs363006, rs1051312, rs8636, rs362549, and rs362998) of SNAP25 and ADHD in a pooled sample of ten family-based studies and four case-control studies by using meta-analysis. The combined analysis results were significant only for rs3746544 (P = 0.010) with mild association (odds ratio (OR) = 1.14). And, the meta-analysis data for rs8636, rs362549, and rs362998 are the first time to be reported; however, no positive association was detected. In conclusion, we report some evidence supporting the association of SNAP25 to ADHD. Future research should emphasize genome-wide association studies in more specific subgroups and larger independent samples.
Collapse
Affiliation(s)
- Yun-Sheng Liu
- Key Laboratory of Environment and Health, Ministry of Education & Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Xuan Dai
- Key Laboratory of Environment and Health, Ministry of Education & Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Wei Wu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Fang-Fen Yuan
- Key Laboratory of Environment and Health, Ministry of Education & Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Xue Gu
- Key Laboratory of Environment and Health, Ministry of Education & Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Jian-Guo Chen
- Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.,Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Ling-Qiang Zhu
- Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China. .,Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Jing Wu
- Key Laboratory of Environment and Health, Ministry of Education & Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
21
|
Cupertino RB, Kappel DB, Bandeira CE, Schuch JB, da Silva BS, Müller D, Bau CHD, Mota NR. SNARE complex in developmental psychiatry: neurotransmitter exocytosis and beyond. J Neural Transm (Vienna) 2016; 123:867-83. [DOI: 10.1007/s00702-016-1514-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 01/20/2016] [Indexed: 12/31/2022]
|
22
|
Kosiorek M, Podszywalow-Bartnicka P, Zylinska L, Pikula S. NFAT1 and NFAT3 cooperate with HDAC4 during regulation of alternative splicing of PMCA isoforms in PC12 cells. PLoS One 2014; 9:e99118. [PMID: 24905014 PMCID: PMC4048221 DOI: 10.1371/journal.pone.0099118] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 05/10/2014] [Indexed: 02/07/2023] Open
Abstract
Background The bulk of human genes undergo alternative splicing (AS) upon response to physiological stimuli. AS is a great source of protein diversity and biological processes and is associated with the development of many diseases. Pheochromocytoma is a neuroendocrine tumor, characterized by an excessive Ca2+-dependent secretion of catecholamines. This underlines the importance of balanced control of calcium transport via regulation of gene expression pattern, including different calcium transport systems, such as plasma membrane Ca2+-ATPases (PMCAs), abundantly expressed in pheochromocytoma chromaffin cells (PC12 cells). PMCAs are encoded by four genes (Atp2b1, Atp2b2, Atp2b3, Atp2b4), whose transcript products undergo alternative splicing giving almost 30 variants. Results In this scientific report, we propose a novel mechanism of regulation of PMCA alternative splicing in PC12 cells through cooperation of the nuclear factor of activated T-cells (NFAT) and histone deacetylases (HDACs). Luciferase assays showed increased activity of NFAT in PC12 cells, which was associated with altered expression of PMCA. RT-PCR experiments suggested that inhibition of the transcriptional activity of NFAT might result in the rearrangement of PMCA splicing variants in PC12 cells. NFAT inhibition led to dominant expression of 2x/c, 3x/a and 4x/a PMCA variants, while in untreated cells the 2w,z/b, 3z,x/b,c,e,f, and 4x/b variants were found as well. Furthermore, chromatin immunoprecipitation experiments showed that NFAT1-HDAC4 or NFAT3-HDAC4 complexes might be involved in regulation of PMCA2x splicing variant generation. Conclusions We suggest that the influence of NFAT/HDAC on PMCA isoform composition might be important for altered dopamine secretion by PC12 cells.
Collapse
Affiliation(s)
- Michalina Kosiorek
- Department of Biochemistry, Nencki Institute of Experimental Biology, Warsaw, Poland
- Department of Neurodegenerative Disorders, Laboratory of Neurogenetics, Mossakowski Medical Research Centre PAS, Warsaw, Poland
| | | | - Ludmila Zylinska
- Department of Molecular Neurochemistry, Medical University, Lodz, Poland
| | - Slawomir Pikula
- Department of Biochemistry, Nencki Institute of Experimental Biology, Warsaw, Poland
- * E-mail:
| |
Collapse
|
23
|
Wang W, Wang F, Liu J, Zhao W, Zhao Q, He M, Qian BJ, Xu Y, Liu R, Liu SJ, Liu W, Liu J, Zhou XF, Wang TH. SNAP25 ameliorates sensory deficit in rats with spinal cord transection. Mol Neurobiol 2014; 50:290-304. [PMID: 24519330 DOI: 10.1007/s12035-014-8642-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 01/03/2014] [Indexed: 02/05/2023]
Abstract
Spinal cord injury causes sensory loss below the level of lesion. Synaptosomal-associated protein 25 (SNAP25) is a t-SNARE protein essential for exocytosis and neurotransmitter release, but its role in sensory functional recovery has not been determined. The aim of the present study is therefore to investigate whether SNAP25 can promote sensory recovery. By 2D proteomics, we found a downregulation of SNAP25 and then constructed two lentiviral vectors, Lv-exSNAP25 and Lv-shSNAP25, which allows efficient and stable RNAi-mediated silencing of endogenous SNAP25. Overexpression of SNAP25 enhanced neurite outgrowth in vitro and behavior response to thermal and mechanical stimuli in vivo, while the silencing of SNAP25 had the opposite effect. These results suggest that SNAP25 plays a crucial role in sensory functional recovery following spinal cord injury (SCI). Our study therefore provides a novel target for the management of SCI for sensory dysfunction.
Collapse
Affiliation(s)
- Wei Wang
- Department of Anesthesiology and Institute of Neurological Disease, Translation Neuroscience Center, The State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Synaptotagmin interaction with SNAP-25 governs vesicle docking, priming, and fusion triggering. J Neurosci 2013; 33:14417-30. [PMID: 24005294 DOI: 10.1523/jneurosci.1236-13.2013] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
SNARE complex assembly constitutes a key step in exocytosis that is rendered Ca(2+)-dependent by interactions with synaptotagmin-1. Two putative sites for synaptotagmin binding have recently been identified in SNAP-25 using biochemical methods: one located around the center and another at the C-terminal end of the SNARE bundle. However, it is still unclear whether and how synaptotagmin-1 × SNARE interactions at these sites are involved in regulating fast neurotransmitter release. Here, we have used electrophysiological techniques with high time-resolution to directly investigate the mechanistic ramifications of proposed SNAP-25 × synaptotagmin-1 interaction in mouse chromaffin cells. We demonstrate that the postulated central binding domain surrounding layer zero covers both SNARE motifs of SNAP-25 and is essential for vesicle docking, priming, and fast fusion-triggering. Mutation of this site caused no further functional alterations in synaptotagmin-1-deficient cells, indicating that the central acidic patch indeed constitutes a mechanistically relevant synaptotagmin-1 interaction site. Moreover, our data show that the C-terminal binding interface only plays a subsidiary role in triggering but is required for the full size of the readily releasable pool. Intriguingly, we also found that mutation of synaptotagmin-1 interaction sites led to more pronounced phenotypes in the context of the adult neuronal isoform SNAP-25B than in the embryonic isoform SNAP-25A. Further experiments demonstrated that stronger synaptotagmin-1 × SNAP-25B interactions allow for the larger primed vesicle pool supported by SNAP-25 isoform B. Thus, synaptotagmin-1 × SNARE interactions are not only required for multiple mechanistic steps en route to fusion but also underlie the developmental control of the releasable vesicle pool.
Collapse
|
25
|
Yilmaz M, Edgunlu TG, Yilmaz N, Cetin ES, Celik SK, Emir GK, Sözen A. Genetic variants of synaptic vesicle and presynaptic plasma membrane proteins in idiopathic generalized epilepsy. J Recept Signal Transduct Res 2013; 34:38-43. [PMID: 24164654 DOI: 10.3109/10799893.2013.848893] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND The aim of this study was to analyze the role of the genetic variants of two synaptic vesicle proteins (VAMP2 and Synaptotagmin XI) and two presynaptic plasma membrane proteins (Syntaxin 1A and SNAP-25) in patients with idiopathic generalized epilepsy (IGE). METHOD Eighty-five patients with IGE and 93 healthy subjects were included in the study. We analyzed the functional polymorphisms of VAMP2, Synaptotagmin XI, Syntaxin 1A and SNAP-25 genes with polymerase chain reaction and restriction fragment length polymorphism methods. RESULTS In the patients with IGE, significant differences alleles and genotypes of 26 bp Ins/Del polymorphism of the VAMP2 gene and the 33-bp promoter region of Synaptotagmin XI were observed, however no associaton was found regarding Intron 7 rs1569061 of Syntaxin 1A gene, MnlI rs3746544 and DdeI rs1051312 polymorphisms of SNAP-25 gene compared with healthy subjects. Carriers of the C allele of Synaptotagmin XI had worse measures compared with the T allele of Synaptotagmin XI. In the haplotype analysis, the frequency of the T alleles of rs1569061 and of the C alleles of the 33-bp promoter region of Synaptotagmin XI was found to be significantly higher in patients with IGE as compared with the healthy subjects. CONCLUSION The genetic variations of VAMP2, Synaptotagmin XI might be indication of the relationship between these genes and IGE.
Collapse
|
26
|
Kelemen O, Convertini P, Zhang Z, Wen Y, Shen M, Falaleeva M, Stamm S. Function of alternative splicing. Gene 2013; 514:1-30. [PMID: 22909801 PMCID: PMC5632952 DOI: 10.1016/j.gene.2012.07.083] [Citation(s) in RCA: 548] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 07/21/2012] [Accepted: 07/30/2012] [Indexed: 12/15/2022]
Abstract
Almost all polymerase II transcripts undergo alternative pre-mRNA splicing. Here, we review the functions of alternative splicing events that have been experimentally determined. The overall function of alternative splicing is to increase the diversity of mRNAs expressed from the genome. Alternative splicing changes proteins encoded by mRNAs, which has profound functional effects. Experimental analysis of these protein isoforms showed that alternative splicing regulates binding between proteins, between proteins and nucleic acids as well as between proteins and membranes. Alternative splicing regulates the localization of proteins, their enzymatic properties and their interaction with ligands. In most cases, changes caused by individual splicing isoforms are small. However, cells typically coordinate numerous changes in 'splicing programs', which can have strong effects on cell proliferation, cell survival and properties of the nervous system. Due to its widespread usage and molecular versatility, alternative splicing emerges as a central element in gene regulation that interferes with almost every biological function analyzed.
Collapse
Affiliation(s)
- Olga Kelemen
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Paolo Convertini
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Zhaiyi Zhang
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Yuan Wen
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Manli Shen
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Marina Falaleeva
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Stefan Stamm
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| |
Collapse
|
27
|
|
28
|
Involvement of gecko SNAP25b in spinal cord regeneration by promoting outgrowth and elongation of neurites. Int J Biochem Cell Biol 2012; 44:2288-98. [DOI: 10.1016/j.biocel.2012.09.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 09/12/2012] [Accepted: 09/14/2012] [Indexed: 01/17/2023]
|
29
|
Upreti C, Zhang XL, Alford S, Stanton PK. Role of presynaptic metabotropic glutamate receptors in the induction of long-term synaptic plasticity of vesicular release. Neuropharmacology 2012; 66:31-9. [PMID: 22626985 DOI: 10.1016/j.neuropharm.2012.05.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 05/07/2012] [Accepted: 05/09/2012] [Indexed: 11/24/2022]
Abstract
While postsynaptic ionotropic and metabotropic glutamate receptors have received the lions share of attention in studies of long-term activity-dependent synaptic plasticity, it is becoming clear that presynaptic metabotropic glutamate receptors play critical roles in both short-term and long-term plasticity of vesicular transmitter release, and that they act both at the level of voltage-dependent calcium channels and directly on proteins of the vesicular release machinery. Activation of G protein-coupled receptors can transiently inhibit vesicular release through the release of Gβγ which binds to both voltage-dependent calcium channels to reduce calcium influx, and directly to the C-terminus region of the SNARE protein SNAP-25. Our recent work has revealed that the binding of Gβγ to SNAP-25 is necessary, but not sufficient, to elicit long-term depression (LTD) of vesicular glutamate release, and that the concomitant release of Gα(i) and the second messenger nitric oxide are also necessary steps in the presynaptic LTD cascade. Here, we review the current state of knowledge of the molecular steps mediating short-term and long-term plasticity of vesicular release at glutamatergic synapses, and the many gaps that remain to be addressed. This article is part of a Special Issue entitled 'Metabotropic Glutamate Receptors'.
Collapse
Affiliation(s)
- Chirag Upreti
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY 10595, USA
| | | | | | | |
Collapse
|
30
|
Mohrmann R, Sørensen JB. SNARE requirements en route to exocytosis: from many to few. J Mol Neurosci 2012; 48:387-94. [PMID: 22427188 DOI: 10.1007/s12031-012-9744-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 02/29/2012] [Indexed: 12/30/2022]
Abstract
Although it has been known for almost two decades that the ternary complex of N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) constitutes the functional unit driving membrane fusion, our knowledge about the dynamical arrangement and organization of SNARE proteins and their complexes before and during vesicle exocytosis is still limited. Here, we review recent progress in this expanding field with emphasis on the question of fusion complex stoichiometry, i.e., how many SNARE proteins and complexes are needed for the fusion of a vesicle with the plasma membrane.
Collapse
Affiliation(s)
- Ralf Mohrmann
- Department of Physiology, University of Saarland, Homburg, Germany.
| | | |
Collapse
|
31
|
Scullin CS, Tafoya LC, Wilson MC, Partridge LD. Presynaptic residual calcium and synaptic facilitation at hippocampal synapses of mice with altered expression of SNAP-25. Brain Res 2012; 1431:1-12. [PMID: 22119397 PMCID: PMC3246534 DOI: 10.1016/j.brainres.2011.10.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Revised: 10/19/2011] [Accepted: 10/20/2011] [Indexed: 10/15/2022]
Abstract
Paired pulse facilitation (PPF) is a form of short-term synaptic plasticity that results from an interaction of residual presynaptic Ca(2+) ([Ca(2+)](res)), number of release-competent vesicles, and the sensitivity of the vesicle release mechanisms to Ca(2+). While PPF is predominant at hippocampal Schaffer collateral-CA1 (SC-CA1) synapses, facilitation is greater in adult mice (designated Tkneo) that over express an isoform of the plasma membrane-targeted SNARE protein, SNAP-25a, which is normally predominantly expressed in juvenile animals. SNAP-25 is essential for action potential-dependent neuroexocytosis, yet the significance of the shift between the alternatively spliced variants SNAP-25a and SNAP-25b is not fully understood. This alteration of a key component of the protein machinery required for neurotransmitter release in Tkneo mice, therefore, provides a useful tool to further investigate presynaptic mechanisms that influence short-term plasticity. To explore this link between SNAP-25 and PPF, we simultaneously measured postsynaptic potentials and presynaptic [Ca(2+)](res) during paired-pulses in adult Tkneo, heterozygote null (HET), and wild type (WT) mice. We demonstrate that enhanced PPF is maintained at mature hippocampal synapses of Tkneo mice that predominantly express SNAP-25a, and that [Ca(2+)](res) kinetics are altered at synapses of Tkneo and HET mice, both of which exhibit reduced levels of total SNAP-25 expression. To evaluate the role of SNAP-25 in short-term plasticity and [Ca(2+)](res) regulation, we applied a vesicular release probability model for neurotransmission. Our results suggest that the isoform expression and total level of SNAP-25 affect both [Ca(2+)](res) dynamics and the ability of releasable vesicles to enter into a facilitated state.
Collapse
Affiliation(s)
- Chessa S. Scullin
- Department of Neurosciences, University of New Mexico, Albuquerque, NM 87131
| | - Lawrence C. Tafoya
- Department of Neurosciences, University of New Mexico, Albuquerque, NM 87131
| | - Michael C. Wilson
- Department of Neurosciences, University of New Mexico, Albuquerque, NM 87131
| | - L. Donald Partridge
- Department of Neurosciences, University of New Mexico, Albuquerque, NM 87131
| |
Collapse
|
32
|
Gβγ and the C terminus of SNAP-25 are necessary for long-term depression of transmitter release. PLoS One 2011; 6:e20500. [PMID: 21633701 PMCID: PMC3102109 DOI: 10.1371/journal.pone.0020500] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Accepted: 05/04/2011] [Indexed: 11/19/2022] Open
Abstract
Background Short-term presynaptic inhibition mediated by G protein-coupled receptors involves a direct interaction between G proteins and the vesicle release machinery. Recent studies implicate the C terminus of the vesicle-associated protein SNAP-25 as a molecular binding target of Gβγ that transiently reduces vesicular release. However, it is not known whether SNAP-25 is a target for molecular modifications expressing long-term changes in transmitter release probability. Methodology/Principal Findings This study utilized two-photon laser scanning microscopy for real-time imaging of action potential-evoked [Ca2+] increases, in single Schaffer collateral presynaptic release sites in in vitro hippocampal slices, plus simultaneous recording of Schaffer collateral-evoked synaptic potentials. We used electroporation to infuse small peptides through CA3 cell bodies into presynaptic Schaffer collateral terminals to selectively study the presynaptic effect of scavenging the G-protein Gβγ. We demonstrate here that the C terminus of SNAP-25 is necessary for expression of LTD, but not long-term potentiation (LTP), of synaptic strength. Using type A botulinum toxin (BoNT/A) to enzymatically cleave the 9 amino acid C-terminus of SNAP-25 eliminated the ability of low frequency synaptic stimulation to induce LTD, but not LTP, even if release probability was restored to pre-BoNT/A levels by elevating extracellular [Ca2+]. Presynaptic electroporation infusion of the 14-amino acid C-terminus of SNAP-25 (Ct-SNAP-25), to scavenge Gβγ, reduced both the transient presynaptic inhibition produced by the group II metabotropic glutamate receptor stimulation, and LTD. Furthermore, presynaptic infusion of mSIRK, a second, structurally distinct Gβγ scavenging peptide, also blocked the induction of LTD. While Gβγ binds directly to and inhibit voltage-dependent Ca2+ channels, imaging of presynaptic [Ca2+] with Mg-Green revealed that low-frequency stimulation only transiently reduced presynaptic Ca2+ influx, an effect not altered by infusion of Ct-SNAP-25. Conclusions/Significance The C-terminus of SNAP-25, which links synaptotagmin I to the SNARE complex, is a binding target for Gβγ necessary for both transient transmitter-mediated presynaptic inhibition, and the induction of presynaptic LTD.
Collapse
|
33
|
Vacca M, Albania L, Della Ragione F, Carpi A, Rossi V, Strazzullo M, De Franceschi N, Rossetto O, Filippini F, D'Esposito M. Alternative splicing of the human gene SYBL1 modulates protein domain architecture of Longin VAMP7/TI-VAMP, showing both non-SNARE and synaptobrevin-like isoforms. BMC Mol Biol 2011; 12:26. [PMID: 21609427 PMCID: PMC3123573 DOI: 10.1186/1471-2199-12-26] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 05/24/2011] [Indexed: 11/15/2022] Open
Abstract
Background The control of intracellular vesicle trafficking is an ideal target to weigh the role of alternative splicing in shaping genomes to make cells. Alternative splicing has been reported for several Soluble N-ethylmaleimide-sensitive factor Attachment protein REceptors of the vesicle (v-SNAREs) or of the target membrane (t-SNARES), which are crucial to intracellular membrane fusion and protein and lipid traffic in Eukaryotes. However, splicing has not yet been investigated in Longins, i.e. the most widespread v-SNAREs. Longins are essential in Eukaryotes and prototyped by VAMP7, Sec22b and Ykt6, sharing a conserved N-terminal Longin domain which regulates membrane fusion and subcellular targeting. Human VAMP7/TI-VAMP, encoded by gene SYBL1, is involved in multiple cell pathways, including control of neurite outgrowth. Results Alternative splicing of SYBL1 by exon skipping events results in the production of a number of VAMP7 isoforms. In-frame or frameshift coding sequence modifications modulate domain architecture of VAMP7 isoforms, which can lack whole domains or domain fragments and show variant or extra domains. Intriguingly, two main types of VAMP7 isoforms either share the inhibitory Longin domain and lack the fusion-promoting SNARE motif, or vice versa. Expression analysis in different tissues and cell lines, quantitative real time RT-PCR and confocal microscopy analysis of fluorescent protein-tagged isoforms demonstrate that VAMP7 variants have different tissue specificities and subcellular localizations. Moreover, design and use of isoform-specific antibodies provided preliminary evidence for the existence of splice variants at the protein level. Conclusions Previous evidence on VAMP7 suggests inhibitory functions for the Longin domain and fusion/growth promoting activity for the Δ-longin molecule. Thus, non-SNARE isoforms with Longin domain and non-longin SNARE isoforms might have somehow opposite regulatory functions. When considering splice variants as "natural mutants", evidence on modulation of subcellular localization by variation in domain combination can shed further light on targeting determinants. Although further work will be needed to characterize identified variants, our data might open the route to unravel novel molecular partners and mechanisms, accounting for the multiplicity of functions carried out by the different members of the Longin proteins family.
Collapse
Affiliation(s)
- Marcella Vacca
- Institute of Genetics and Biophysics A.Buzzati Traverso Consiglio Nazionale delle Ricerche, via P. Castellino 111, 80131 Naples, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Hirano AA, Brandstätter JH, Morgans CW, Brecha NC. SNAP25 expression in mammalian retinal horizontal cells. J Comp Neurol 2011; 519:972-88. [PMID: 21280047 DOI: 10.1002/cne.22562] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Horizontal cells mediate inhibitory feedforward and feedback lateral interactions in the outer retina at photoreceptor terminals and bipolar cell dendrites; however, the mechanisms that underlie synaptic transmission from mammalian horizontal cells are poorly understood. The localization of a vesicular γ-aminobutyric acid (GABA) transporter (VGAT) to horizontal cell processes in primate and rodent retinae suggested that mammalian horizontal cells release transmitter in a vesicular manner. Toward determining whether the molecular machinery for vesicular transmitter release is present in horizontal cells, we investigated the expression of SNAP25 (synaptosomal-associated protein of 25 kDa), a key SNARE protein, by immunocytochemistry with cell type-specific markers in the retinae of mouse, rat, rabbit, and monkey. Different commercial antibodies to SNAP25 were tested on vertical sections of retina. We report the robust expression of SNAP25 in both plexiform layers. Double labeling with SNAP25 and calbindin antibodies demonstrated that horizontal cell processes and their endings in photoreceptor triad synapses were strongly labeled for both proteins in mouse, rat, rabbit, and monkey retinae. Double labeling with parvalbumin antibodies in monkey retina verified SNAP25 immunoreactivity in all horizontal cells. Pre-embedding immunoelectron microscopy in rabbit retina confirmed expression of SNAP25 in lateral elements within photoreceptor triad synapses. The SNAP25 immunoreactivity in the plexiform layers and outer nuclear layer fell into at least three patterns depending on the antibody, suggesting a differential distribution of SNAP25 isoforms. The presence of SNAP25a and SNAP25b isoforms in mouse retina was established by reverse transcriptase-polymerase chain reaction. SNAP25 expression in mammalian horizontal cells along with other SNARE proteins is consistent with vesicular exocytosis.
Collapse
Affiliation(s)
- Arlene A Hirano
- Department of Neurobiology, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California 90095, USA.
| | | | | | | |
Collapse
|
35
|
Prescott GR, Chamberlain LH. Regional and developmental brain expression patterns of SNAP25 splice variants. BMC Neurosci 2011; 12:35. [PMID: 21526988 PMCID: PMC3104942 DOI: 10.1186/1471-2202-12-35] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Accepted: 04/28/2011] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND SNAP25 is an essential SNARE protein for regulated exocytosis in neuronal cells. Differential splicing of the SNAP25 gene results in the expression of two transcripts, SNAP25a and SNAP25b. These splice variants differ by only 9 amino acids, and studies of their expression to date have been limited to analysis of the corresponding mRNAs. Although these studies have been highly informative, it is possible that factors such as differential turnover of the SNAP25 proteins could complicate interpretations based entirely on mRNA expression profiles. RESULTS We report the generation and characterization of antibodies that distinguish between SNAP25a and SNAP25b isoforms, and their use to investigate the expression profile of these proteins in rat and human brain. In rat brain, SNAP25b protein expression increased dramatically during post-natal development, whereas the increase in SNAP25a expression was more modest and variable. The extent of this up-regulation in SNAP25b expression was similar across cortex, cerebellum and hippocampus. The SNAP25 isoforms also displayed distinct regional expression patterns, with SNAP25a very weakly expressed in both rat and human cerebellum. Quantitative analysis revealed that SNAP25b was the dominant isoform in all adult human brain regions examined. CONCLUSIONS SNAP25a and SNAP25b display distinct developmental and regional expression profiles in rat and human brain. These differences might reflect distinct functions of these highly conserved isoforms in membrane fusion pathways in the brain. The antibodies generated and characterized in this study represent important tools for future analyses of these essential SNARE protein isoforms.
Collapse
Affiliation(s)
- Gerald R Prescott
- Centre for Integrative Physiology, School of Biomedical Sciences, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Luke H Chamberlain
- Centre for Integrative Physiology, School of Biomedical Sciences, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, UK
| |
Collapse
|
36
|
Ca2+ induces clustering of membrane proteins in the plasma membrane via electrostatic interactions. EMBO J 2011; 30:1209-20. [PMID: 21364530 DOI: 10.1038/emboj.2011.53] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Accepted: 02/02/2011] [Indexed: 12/23/2022] Open
Abstract
Membrane proteins and membrane lipids are frequently organized in submicron-sized domains within cellular membranes. Factors thought to be responsible for domain formation include lipid-lipid interactions, lipid-protein interactions and protein-protein interactions. However, it is unclear whether the domain structure is regulated by other factors such as divalent cations. Here, we have examined in native plasma membranes and intact cells the role of the second messenger Ca(2+) in membrane protein organization. We find that Ca(2+) at low micromolar concentrations directly redistributes a structurally diverse array of membrane proteins via electrostatic effects. Redistribution results in a more clustered pattern, can be rapid and triggered by Ca(2+) influx through voltage-gated calcium channels and is reversible. In summary, the data demonstrate that the second messenger Ca(2+) strongly influences the organization of membrane proteins, thus adding a novel and unexpected factor that may control the domain structure of biological membranes.
Collapse
|
37
|
Etain B, Dumaine A, Mathieu F, Chevalier F, Henry C, Kahn JP, Deshommes J, Bellivier F, Leboyer M, Jamain S. A SNAP25 promoter variant is associated with early-onset bipolar disorder and a high expression level in brain. Mol Psychiatry 2010; 15:748-55. [PMID: 19125158 PMCID: PMC2937032 DOI: 10.1038/mp.2008.148] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Bipolar disorder (BD) is one of the most common and persistent psychiatric disorders. Early-onset BD has been shown to be the most severe and familial form. We recently carried out a whole-genome linkage analysis on sibpairs affected by early-onset BD and showed that the 20p12 region was more frequently shared in our families than expected by chance. The synaptosomal-associated protein SNAP25 is a presynaptic plasma membrane protein essential for the triggering of vesicular fusion and neurotransmitter release, and for which abnormal protein levels have been reported in postmortem studies of bipolar patients. We hypothesised that variations in the gene encoding SNAP25, located on chromosome 20p12, might influence the susceptibility to early-onset BD. We screened SNAP25 for mutations and performed a case-control association study in 197 patients with early-onset BD, 202 patients with late-onset BD and 136 unaffected subjects. In addition, we analysed the expression level of the two SNAP25 isoforms in 60 brains. We showed that one variant, located in the promoter region, was associated with early-onset BD but not with the late-onset subgroup. In addition, individuals homozygous for this variant showed a significant higher SNAP25b expression level in prefrontal cortex. These results show that variations in SNAP25, associated with an increased gene expression level in prefrontal cortex, might predispose to early-onset BD. Further analyses of this gene, as well as analysis of genes encoding for the SNAP25 protein partners, are required to understand the impact of such molecular mechanisms in BD.
Collapse
Affiliation(s)
- Bruno Etain
- Institut Mondor de Recherche Biomédicale
INSERM : U955Université Paris XII Val de MarneIFR10FR,Pôle de psychiatrie
AP-HPGroupe Henri Mondor-Albert ChenevierCréteil,FR
| | - Anne Dumaine
- Institut Mondor de Recherche Biomédicale
INSERM : U955Université Paris XII Val de MarneIFR10FR
| | - Flavie Mathieu
- Institut Mondor de Recherche Biomédicale
INSERM : U955Université Paris XII Val de MarneIFR10FR
| | - Fabien Chevalier
- Institut Mondor de Recherche Biomédicale
INSERM : U955Université Paris XII Val de MarneIFR10FR
| | - Chantal Henry
- Institut Mondor de Recherche Biomédicale
INSERM : U955Université Paris XII Val de MarneIFR10FR,Pôle de psychiatrie
AP-HPGroupe Henri Mondor-Albert ChenevierCréteil,FR
| | - Jean-Pierre Kahn
- Service de psychiatrie et psychologie clinique
CHU NancyHôpital Jeanne-d'ArcNancy,FR
| | - Jasmine Deshommes
- Institut Mondor de Recherche Biomédicale
INSERM : U955Université Paris XII Val de MarneIFR10FR,Pôle de psychiatrie
AP-HPGroupe Henri Mondor-Albert ChenevierCréteil,FR
| | - Frank Bellivier
- Institut Mondor de Recherche Biomédicale
INSERM : U955Université Paris XII Val de MarneIFR10FR,Pôle de psychiatrie
AP-HPGroupe Henri Mondor-Albert ChenevierCréteil,FR
| | - Marion Leboyer
- Institut Mondor de Recherche Biomédicale
INSERM : U955Université Paris XII Val de MarneIFR10FR,Pôle de psychiatrie
AP-HPGroupe Henri Mondor-Albert ChenevierCréteil,FR
| | - Stéphane Jamain
- Institut Mondor de Recherche Biomédicale
INSERM : U955Université Paris XII Val de MarneIFR10FR,* Correspondence should be adressed to: Stéphane Jamain
| |
Collapse
|
38
|
Halemani ND, Bethani I, Rizzoli SO, Lang T. Structure and Dynamics of a Two-Helix SNARE Complex in Live Cells. Traffic 2010; 11:394-404. [DOI: 10.1111/j.1600-0854.2009.01020.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
39
|
|
40
|
Johansson JU, Ericsson J, Janson J, Beraki S, Stanić D, Mandic SA, Wikström MA, Hökfelt T, Ögren SO, Rozell B, Berggren PO, Bark C. An ancient duplication of exon 5 in the Snap25 gene is required for complex neuronal development/function. PLoS Genet 2008; 4:e1000278. [PMID: 19043548 PMCID: PMC2581893 DOI: 10.1371/journal.pgen.1000278] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Accepted: 10/27/2008] [Indexed: 01/05/2023] Open
Abstract
Alternative splicing is an evolutionary innovation to create functionally diverse proteins from a limited number of genes. SNAP-25 plays a central role in neuroexocytosis by bridging synaptic vesicles to the plasma membrane during regulated exocytosis. The SNAP-25 polypeptide is encoded by a single copy gene, but in higher vertebrates a duplication of exon 5 has resulted in two mutually exclusive splice variants, SNAP-25a and SNAP-25b. To address a potential physiological difference between the two SNAP-25 proteins, we generated gene targeted SNAP-25b deficient mouse mutants by replacing the SNAP-25b specific exon with a second SNAP-25a equivalent. Elimination of SNAP-25b expression resulted in developmental defects, spontaneous seizures, and impaired short-term synaptic plasticity. In adult mutants, morphological changes in hippocampus and drastically altered neuropeptide expression were accompanied by severe impairment of spatial learning. We conclude that the ancient exon duplication in the Snap25 gene provides additional SNAP-25-function required for complex neuronal processes in higher eukaryotes. In evolution, duplication of genes or gene segments appears to be an efficient way to add diverse functions in more complex organisms. The SNAP-25 protein plays an important role in mediating the release of neurotransmitters and hormones. SNAP-25 exists as two variants: SNAP-25a, which is present in early development, and SNAP-25b, which is most abundant from early adulthood and onwards. We have developed mouse mutants that only express SNAP-25a, but retain normal SNAP-25 levels by replacing the SNAP-25b segment in the Snap25 gene with an additional SNAP-25a copy. We show that SNAP-25b is required for early postnatal development and that a balanced expression of the two proteins is a prerequisite for maintaining an operational neuronal network during adulthood. Mice that only have SNAP-25a develop seizures, and show learning deficits and anxiety. Synaptic plasticity is impaired, and structural changes are observed in areas that are connected to such behavioral functions. In man, SNAP-25 function has been linked to behavioral and neuropsychiatric disorders, including attention deficit hyperactivity disorder, ADHD. Our present findings using genetic elimination of SNAP-25b suggest that even small alterations in the regulation of the Snap25 gene, resulting in a disturbed balance between SNAP-25a and SNAP-25b, lead to nervous system dysfunction.
Collapse
Affiliation(s)
- Jenny U. Johansson
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Jesper Ericsson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Juliette Janson
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Simret Beraki
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Davor Stanić
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Slavena A. Mandic
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | | | - Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Sven Ove Ögren
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Björn Rozell
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Christina Bark
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
41
|
The role of the t-SNARE SNAP-25 in action potential-dependent calcium signaling and expression in GABAergic and glutamatergic neurons. BMC Neurosci 2008; 9:105. [PMID: 18959796 PMCID: PMC2600647 DOI: 10.1186/1471-2202-9-105] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2008] [Accepted: 10/29/2008] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND The soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex, comprised of SNAP-25, syntaxin 1A, and VAMP-2, has been shown to be responsible for action potential (AP)-dependent, calcium-triggered release of several neurotransmitters. However, this basic fusogenic protein complex may be further specialized to suit the requirements for different neurotransmitter systems, as exemplified by neurons and neuroendocrine cells. In this study, we investigate the effects of SNAP-25 ablation on spontaneous neuronal activity and the expression of functionally distinct isoforms of this t-SNARE in GABAergic and glutamatergic neurons of the adult brain. RESULTS We found that neurons cultured from Snap25 homozygous null mutant (Snap25-/-) mice failed to develop synchronous network activity seen as spontaneous AP-dependent calcium oscillations and were unable to trigger glial transients following depolarization. Voltage-gated calcium channel (VGCC) mediated calcium transients evoked by depolarization, nevertheless, did not differ between soma of SNAP-25 deficient and control neurons. Furthermore, we observed that although the expression of SNAP-25 RNA transcripts varied among neuronal populations in adult brain, the relative ratio of the transcripts encoding alternatively spliced SNAP-25 variant isoforms was not different in GABAergic and glutamatergic neurons. CONCLUSION We propose that the SNAP-25b isoform is predominantly expressed by both mature glutamatergic and GABAergic neurons and serves as a fundamental component of SNARE complex used for fast synaptic communication in excitatory and inhibitory circuits required for brain function. Moreover, SNAP-25 is required for neurons to establish AP-evoked synchronous network activity, as measured by calcium transients, whereas the loss of this t-SNARE does not affect voltage-dependent calcium entry.
Collapse
|
42
|
Nagy G, Milosevic I, Mohrmann R, Wiederhold K, Walter AM, Sørensen JB. The SNAP-25 linker as an adaptation toward fast exocytosis. Mol Biol Cell 2008; 19:3769-81. [PMID: 18579690 DOI: 10.1091/mbc.e07-12-1218] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The assembly of four soluble N-ethylmaleimide-sensitive factor attachment protein receptor domains into a complex is essential for membrane fusion. In most cases, the four SNARE-domains are encoded by separate membrane-targeted proteins. However, in the exocytotic pathway, two SNARE-domains are present in one protein, connected by a flexible linker. The significance of this arrangement is unknown. We characterized the role of the linker in SNAP-25, a neuronal SNARE, by using overexpression techniques in synaptosomal-associated protein of 25 kDa (SNAP-25) null mouse chromaffin cells and fast electrophysiological techniques. We confirm that the palmitoylated linker-cysteines are important for membrane association. A SNAP-25 mutant without cysteines supported exocytosis, but the fusion rate was slowed down and the fusion pore duration prolonged. Using chimeric proteins between SNAP-25 and its ubiquitous homologue SNAP-23, we show that the cysteine-containing part of the linkers is interchangeable. However, a stretch of 10 hydrophobic and charged amino acids in the C-terminal half of the SNAP-25 linker is required for fast exocytosis and in its absence the calcium dependence of exocytosis is shifted toward higher concentrations. The SNAP-25 linker therefore might have evolved as an adaptation toward calcium triggering and a high rate of execution of the fusion process, those features that distinguish exocytosis from other membrane fusion pathways.
Collapse
Affiliation(s)
- Gábor Nagy
- Molecular Mechanism of Exocytosis, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | | | | | | | | | | |
Collapse
|
43
|
Affiliation(s)
- Graham C. R. Ellis-Davies
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, Pennsylvania 19102
| |
Collapse
|
44
|
Abstract
In this report, we present features of the neuronal SNARE complex determined by atomistic molecular dynamics simulations. The results are robust for three models, varying force fields (AMBER and GROMOS) and solvent environment (explicit and implicit). An excellent agreement with experimental findings is observed. The SNARE core complex behaves like a stiff rod, with limited conformational dynamics. An accurate picture of the interactions within the complex emerges with a characteristic pattern of atomic contacts, hydrogen bonds, and salt bridges reinforcing the underlying layer structure. This supports the metaphor of a molecular Velcro strip that has been used by others to describe the neuronal fusion complex. No evidence for directionality in the formation of these interactions was found. Electrostatics largely dominates all interactions, with an acidic surface patch structuring the hydration layers surrounding the complex. The interactions within the four-helix bundle are asymmetric, with the synaptobrevin R-SNARE notably exhibiting an increased rigidity with respect to the three Q-SNARE helices. The interaction patterns we observe provide a new tool for interpreting the impact of mutations on the complex.
Collapse
|
45
|
Zhang HM, Su Q, Luo M. Thyroid hormone regulates the expression of SNAP-25 during rat brain development. Mol Cell Biochem 2007; 307:169-75. [PMID: 17909947 DOI: 10.1007/s11010-007-9596-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Accepted: 08/23/2007] [Indexed: 11/25/2022]
Abstract
Thyroid hormones are major regulators of postnatal brain development. Thyroid hormones act through nuclear receptors to modulate the expression of specific genes in the brain. We have used microarray analysis to identify novel responsive genes in 14-day-old hypothyroid rat brains, and discovered that synaptosomal-associated protein of 25 kDa (SNAP-25) was one of the thyroid hormone-responsive genes. SNAP-25 is a presynaptic plasma membrane protein and an integral component of the vesicle docking and fusion machinery mediating secretion of neurotransmitters and is required for neuritic outgrowth and synaptogenesis. Using microarray analysis we have shown that SNAP-25 was down-regulated in the hypothyroid rat brain compared with the age-matched controls. Real-time RT-PCR and western blotting analysis confirmed that SNAP-25 mRNA and protein levels decreased significantly in the developing hypothyroid rat brain. Our data suggest that in the developing rat brain, SNAP-25 expression is regulated by thyroid hormone, and thyroid hormone deficiency can cause decreased expression of SNAP-25 and this may on some level account for the impaired brain development seen in hypothyroidism.
Collapse
Affiliation(s)
- Hong-Mei Zhang
- Department of Endocrinology, Xin Hua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | | | | |
Collapse
|
46
|
Delgado-Martínez I, Nehring RB, Sørensen JB. Differential abilities of SNAP-25 homologs to support neuronal function. J Neurosci 2007; 27:9380-91. [PMID: 17728451 PMCID: PMC6673127 DOI: 10.1523/jneurosci.5092-06.2007] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The SNAP receptor (SNARE) complex, consisting of synaptosome-associated protein of 25 kDa (SNAP-25), synaptobrevin-2, and syntaxin-1, is involved in synaptic vesicles exocytosis. In addition, SNAP-25 has been implicated in constitutive exocytosis processes required for neurite outgrowth. However, at least three isoforms of SNAP-25 have been reported from neurons: SNAP-23, which is also present in non-neuronal cells, and the two alternative splice variants SNAP-25a and SNAP-25b. Here, we studied the differential ability of these isoforms to support the functions previously broadly ascribed to "SNAP-25." We studied the rescue of snap-25 null neurons in culture with different SNAP-25 homologs. We find that deletion of SNAP-25 leads to strongly reduced neuron survival, and, in the few surviving cells, impaired arborization, reduced spontaneous release, and complete arrest of evoked release. Lentiviral expression of SNAP-25a, SNAP-25b, or SNAP-23 rescued neuronal survival, arborization, amplitude, and frequency of spontaneous events. Also evoked release was rescued by all isoforms, but synchronous release required SNAP-25a/b in both glutamatergic and GABAergic neurons. SNAP-23 supported asynchronous release only, reminiscent of synaptotagmin-1 null neurons. SNAP-25b was superior to SNAP-25a in vesicle priming, resembling the shift to larger releasable vesicle pools that accompanies synaptic maturation. These data demonstrate a differential ability of SNAP-25b, SNAP-25a, and SNAP-23 to support neuronal function.
Collapse
Affiliation(s)
- Ignacio Delgado-Martínez
- Department of Membrane Biophysics, Max Planck Institute for Biophysical Chemistry, D-37077 Göttingen, Germany
| | - Ralf B. Nehring
- Department of Membrane Biophysics, Max Planck Institute for Biophysical Chemistry, D-37077 Göttingen, Germany
| | - Jakob B. Sørensen
- Department of Membrane Biophysics, Max Planck Institute for Biophysical Chemistry, D-37077 Göttingen, Germany
| |
Collapse
|
47
|
Gulyás-Kovács A, de Wit H, Milosevic I, Kochubey O, Toonen R, Klingauf J, Verhage M, Sørensen JB. Munc18-1: sequential interactions with the fusion machinery stimulate vesicle docking and priming. J Neurosci 2007; 27:8676-86. [PMID: 17687045 PMCID: PMC6672934 DOI: 10.1523/jneurosci.0658-07.2007] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2007] [Revised: 06/21/2007] [Accepted: 06/25/2007] [Indexed: 11/21/2022] Open
Abstract
Exocytosis of secretory or synaptic vesicles is executed by a mechanism including the SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) proteins. Munc18-1 is a part of this fusion machinery, but its role is controversial because it is indispensable for fusion but also inhibits the assembly of purified SNAREs in vitro. This inhibition reflects the binding of Munc18-1 to a closed conformation of the target-SNARE syntaxin1. The controversy would be solved if binding to closed syntaxin1 were shown to be stimulatory for vesicle fusion and/or additional essential interactions were identified between Munc18-1 and the fusion machinery. Here, we provide evidence for both notions by dissecting sequential steps of the exocytotic cascade while expressing Munc18 variants in the Munc18-1 null background. In Munc18-1 null chromaffin cells, vesicle docking is abolished and syntaxin levels are reduced. A mutation that diminished Munc18 binding to syntaxin1 in vitro attenuated the vesicle-docking step but rescued vesicle priming in excess of docking. Conversely, expressing the Munc18-2 isoform, which also displays binding to closed syntaxin1, rescued vesicle docking identical with Munc18-1 but impaired more downstream vesicle priming steps. All Munc18 variants restored syntaxin1 levels at least to wild-type levels, showing that the docking phenotype is not caused by syntaxin1 reduction. None of the Munc18 variants affected vesicle fusion kinetics or fusion pore duration. In conclusion, binding of Munc18-1 to closed syntaxin1 stimulates vesicle docking and a distinct interaction mode regulates the consecutive priming step.
Collapse
Affiliation(s)
- Attila Gulyás-Kovács
- Department of Membrane Biophysics, Max Planck Institute for Biophysical Chemistry, D-37077 Göttingen, Germany, and
| | - Heidi de Wit
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam and Vrije Universiteit Medical Center, 1081 HV Amsterdam, The Netherlands
| | - Ira Milosevic
- Department of Membrane Biophysics, Max Planck Institute for Biophysical Chemistry, D-37077 Göttingen, Germany, and
| | - Olexiy Kochubey
- Department of Membrane Biophysics, Max Planck Institute for Biophysical Chemistry, D-37077 Göttingen, Germany, and
| | - Ruud Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam and Vrije Universiteit Medical Center, 1081 HV Amsterdam, The Netherlands
| | - Jürgen Klingauf
- Department of Membrane Biophysics, Max Planck Institute for Biophysical Chemistry, D-37077 Göttingen, Germany, and
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam and Vrije Universiteit Medical Center, 1081 HV Amsterdam, The Netherlands
| | - Jakob B. Sørensen
- Department of Membrane Biophysics, Max Planck Institute for Biophysical Chemistry, D-37077 Göttingen, Germany, and
| |
Collapse
|
48
|
Jeans AF, Oliver PL, Johnson R, Capogna M, Vikman J, Molnár Z, Babbs A, Partridge CJ, Salehi A, Bengtsson M, Eliasson L, Rorsman P, Davies KE. A dominant mutation in Snap25 causes impaired vesicle trafficking, sensorimotor gating, and ataxia in the blind-drunk mouse. Proc Natl Acad Sci U S A 2007; 104:2431-6. [PMID: 17283335 PMCID: PMC1793901 DOI: 10.1073/pnas.0610222104] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Indexed: 01/24/2023] Open
Abstract
The neuronal soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex is essential for synaptic vesicle exocytosis, but its study has been limited by the neonatal lethality of murine SNARE knockouts. Here, we describe a viable mouse line carrying a mutation in the b-isoform of neuronal SNARE synaptosomal-associated protein of 25 kDa (SNAP-25). The causative I67T missense mutation results in increased binding affinities within the SNARE complex, impaired exocytotic vesicle recycling and granule exocytosis in pancreatic beta-cells, and a reduction in the amplitude of evoked cortical excitatory postsynaptic potentials. The mice also display ataxia and impaired sensorimotor gating, a phenotype which has been associated with psychiatric disorders in humans. These studies therefore provide insights into the role of the SNARE complex in both diabetes and psychiatric disease.
Collapse
Affiliation(s)
| | | | | | - Marco Capogna
- Medical Research Council Anatomical Neuropharmacological Unit, University of Oxford, Mansfield Road, Oxford, OX1 3TH, United Kingdom
| | - Jenny Vikman
- Department of Clinical Sciences in Malmö, Clinical Research Centre, Lund University, SE-205 02 Malmö, Sweden; and
| | - Zoltán Molnár
- Department of Physiology, Anatomy, and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, United Kingdom
| | - Arran Babbs
- *Medical Research Council Functional Genetics Unit
| | - Christopher J. Partridge
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, United Kingdom
| | - Albert Salehi
- Department of Clinical Sciences in Malmö, Clinical Research Centre, Lund University, SE-205 02 Malmö, Sweden; and
| | - Martin Bengtsson
- Department of Clinical Sciences in Malmö, Clinical Research Centre, Lund University, SE-205 02 Malmö, Sweden; and
| | - Lena Eliasson
- Department of Clinical Sciences in Malmö, Clinical Research Centre, Lund University, SE-205 02 Malmö, Sweden; and
| | - Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, United Kingdom
| | | |
Collapse
|
49
|
Zilly FE, Sørensen JB, Jahn R, Lang T. Munc18-bound syntaxin readily forms SNARE complexes with synaptobrevin in native plasma membranes. PLoS Biol 2007; 4:e330. [PMID: 17002520 PMCID: PMC1570500 DOI: 10.1371/journal.pbio.0040330] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2006] [Accepted: 08/08/2006] [Indexed: 11/18/2022] Open
Abstract
Munc18-1, a protein essential for regulated exocytosis in neurons and neuroendocrine cells, belongs to the family of Sec1/Munc18-like (SM) proteins. In vitro, Munc18-1 forms a tight complex with the SNARE syntaxin 1, in which syntaxin is stabilized in a closed conformation. Since closed syntaxin is unable to interact with its partner SNAREs SNAP-25 and synaptobrevin as required for membrane fusion, it has hitherto not been possible to reconcile binding of Munc18-1 to syntaxin 1 with its biological function. We now show that in intact and exocytosis-competent lawns of plasma membrane, Munc18-1 forms a complex with syntaxin that allows formation of SNARE complexes. Munc18-1 associated with membrane-bound syntaxin 1 can be effectively displaced by adding recombinant synaptobrevin but not syntaxin 1 or SNAP-25. Displacement requires the presence of endogenous SNAP-25 since no displacement is observed when chromaffin cell membranes from SNAP-25-deficient mice are used. We conclude that Munc18-1 allows for the formation of a complex between syntaxin and SNAP-25 that serves as an acceptor for vesicle-bound synaptobrevin and that thus represents an intermediate in the pathway towards exocytosis.
Collapse
Affiliation(s)
- Felipe E Zilly
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Jakob B Sørensen
- Department of Membrane Biophysics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Reinhard Jahn
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Thorsten Lang
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
50
|
Tafoya LCR, Mameli M, Miyashita T, Guzowski JF, Valenzuela CF, Wilson MC. Expression and function of SNAP-25 as a universal SNARE component in GABAergic neurons. J Neurosci 2006; 26:7826-38. [PMID: 16870728 PMCID: PMC6674219 DOI: 10.1523/jneurosci.1866-06.2006] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Intracellular vesicular trafficking and membrane fusion are important processes for nervous system development and for the function of neural circuits. Synaptosomal-associated protein 25 kDa (SNAP-25) is a component of neural soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) core complexes that mediate the exocytotic release of neurotransmitters at chemical synapses. Previous results from mouse mutant models and pharmacological/neurotoxin blockades have demonstrated a critical role for SNAP-25-containing SNARE complexes in action potential (AP)-dependent release at cholinergic and glutamatergic synapses and for calcium-triggered catecholamine release from chromaffin cells. To examine whether SNAP-25 participates in the evoked release of other neurotransmitters, we investigated the expression and function of SNAP-25 in GABAergic terminals. Patch-clamp recordings in fetal Snap25-null mutant cortex demonstrated that ablation of SNAP-25 eliminated evoked GABA(A) receptor-mediated postsynaptic responses while leaving a low level of spontaneous AP-independent events intact, supporting the involvement of SNAP-25 in the regulated synaptic transmission of early developing GABAergic neurons. In hippocampal cell cultures of wild-type mice, punctate staining of SNAP-25 colocalized with both GABAergic and glutamatergic synaptic markers, whereas stimulus-evoked vesicular recycling was abolished at terminals of both transmitter phenotypes in Snap25-/- neurons. Moreover, immunohistochemistry and fluorescence in situ hybridization revealed coexpression of SNAP-25, VGAT (vesicular GABA transporter), and GAD65/67 (glutamic acid decarboxylase 65/67) in interneurons within several regions of the adult brain. Our results thus provide evidence that SNAP-25 is critical for evoked GABA release during development and is expressed in the presynaptic terminals of mature GABAergic neurons, consistent with its function as a component of a fundamental core SNARE complex required for stimulus-driven neurotransmission.
Collapse
|