1
|
Ruano-Rodríguez S, Navarro-Alonso M, Domínguez-Velasco B, Álvarez-Dolado M, Esteban FJ. STXBP1 Syndrome: Biotechnological Advances, Challenges, and Perspectives in Gene Therapy, Experimental Models, and Translational Research. BIOTECH 2025; 14:11. [PMID: 40227275 PMCID: PMC11939967 DOI: 10.3390/biotech14010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 04/15/2025] Open
Abstract
STXBP1 syndrome is a severe early-onset epileptic encephalopathy characterized by developmental delay and intellectual disability. This review addresses key challenges in STXBP1 syndrome research, focusing on advanced therapeutic approaches and experimental models. We explore gene therapy strategies, including CRISPR-Cas9, adeno-associated viral (AAV) vectors, and RNA therapies such as antisense oligonucleotides (ASOs), aimed at correcting STXBP1 genetic dysfunctions. This review presents in vivo and in vitro models, highlighting their contributions to understanding disease mechanisms. Additionally, we provide a proposal for a detailed bioinformatic analysis of a Spanish cohort of 41 individuals with STXBP1-related disorders, offering insights into specific mutations and their biological implications. Clinical and translational perspectives are discussed, emphasizing the potential of personalized medicine approaches. Future research directions and key challenges are outlined, including the identification of STXBP1 interactors, unexplored molecular pathways, and the need for clinically useful biomarkers. This comprehensive review underscores the complexity of STXBP1-related infantile epileptic encephalopathy and opens new avenues for advancing the understanding and treatment of this heterogeneous disease.
Collapse
Affiliation(s)
- Silvestre Ruano-Rodríguez
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), CSIC-US-JA-UPO, Américo Vespuccio Avenue 24, Cartuja Scientific and Technological Park, 41092 Seville, Spain; (S.R.-R.); (M.N.-A.); (B.D.-V.)
- Systems Biology Unit, Department of Experimental Biology, University of Jaén, Campus Las Lagunillas s/n, 23071 Jaén, Spain
| | - Mar Navarro-Alonso
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), CSIC-US-JA-UPO, Américo Vespuccio Avenue 24, Cartuja Scientific and Technological Park, 41092 Seville, Spain; (S.R.-R.); (M.N.-A.); (B.D.-V.)
| | - Benito Domínguez-Velasco
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), CSIC-US-JA-UPO, Américo Vespuccio Avenue 24, Cartuja Scientific and Technological Park, 41092 Seville, Spain; (S.R.-R.); (M.N.-A.); (B.D.-V.)
| | - Manuel Álvarez-Dolado
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), CSIC-US-JA-UPO, Américo Vespuccio Avenue 24, Cartuja Scientific and Technological Park, 41092 Seville, Spain; (S.R.-R.); (M.N.-A.); (B.D.-V.)
| | - Francisco J. Esteban
- Systems Biology Unit, Department of Experimental Biology, University of Jaén, Campus Las Lagunillas s/n, 23071 Jaén, Spain
| |
Collapse
|
2
|
Tammareddy T, Keyrouz W, Sriram RD, Pant HC, Cardone A, Klauda JB. Investigation of the Effect of Peptide p5 Targeting CDK5-p25 Hyperactivity on Munc18-1 (P67) Regulating Neuronal Exocytosis Using Molecular Simulations. Biochemistry 2024; 63:1837-1857. [PMID: 38953497 DOI: 10.1021/acs.biochem.4c00148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Munc18-1 is an SM (sec1/munc-like) family protein involved in vesicle fusion and neuronal exocytosis. Munc18-1 is known to regulate the exocytosis process by binding with closed- and open-state conformations of Syntaxin1, a protein belonging to the SNARE family established to be central to the exocytosis process. Our previous work studied peptide p5 as a promising drug candidate for CDK5-p25 complex, an Alzheimer's disease (AD) pathological target. Experimental in vivo and in vitro studies suggest that Munc18-1 promotes p5 to selectively inhibit the CDK5-p25 complex without affecting the endogenous CDK5 activity, a characteristic of remarkable therapeutic implications. In this paper, we identify several binding modes of p5 with Munc18-1 that could potentially affect the Munc18-1 binding with SNARE proteins and lead to off-target effects on neuronal communication using molecular dynamics simulations. Recent studies indicate that disruption of Munc18-1 function not only disrupts neurotransmitter release but also results in neurodegeneration, exhibiting clinical resemblance to other neurodegenerative conditions such as AD, causing diagnostic and treatment challenges. We characterize such interactions between p5 and Munc18-1, define the corresponding pharmacophores, and provide guidance for the in vitro validation of our findings to improve therapeutic efficacy and safety of p5.
Collapse
Affiliation(s)
- Tejaswi Tammareddy
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, Maryland 20742, United States
| | | | | | - Harish C Pant
- Neuronal Cytoskeletal Protein Regulation Section, Laboratory of Neurochemistry, NINDS, Bethesda, Maryland 20892, United States
| | | | - Jeffery B Klauda
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, Maryland 20742, United States
- Institute for Physical Science & Technology, Biophysics Graduate Program, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
3
|
Öttl M, Toonen RF, Verhage M. Reduced synaptic depression in human neurons carrying homozygous disease-causing STXBP1 variant L446F. Hum Mol Genet 2024; 33:991-1000. [PMID: 38484778 PMCID: PMC11102591 DOI: 10.1093/hmg/ddae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/02/2024] [Indexed: 05/20/2024] Open
Abstract
MUNC18-1 is an essential protein of the regulated secretion machinery. De novo, heterozygous mutations in STXBP1, the human gene encoding this protein, lead to a severe neurodevelopmental disorder. Here, we describe the electrophysiological characteristics of a unique case of STXBP1-related disorder caused by a homozygous mutation (L446F). We engineered this mutation in induced pluripotent stem cells from a healthy donor (STXBP1LF/LF) to establish isogenic cell models. We performed morphological and electrophysiological analyses on single neurons grown on glial micro-islands. Human STXBP1LF/LF neurons displayed normal morphology and normal basal synaptic transmission but increased paired-pulse ratios and charge released, and reduced synaptic depression compared to control neurons. Immunostainings revealed normal expression levels but impaired recognition by a mutation-specific MUNC18-1 antibody. The electrophysiological gain-of-function phenotype is in line with earlier overexpression studies in Stxbp1 null mouse neurons, with some potentially human-specific features. Therefore, the present study highlights important differences between mouse and human neurons critical for the translatability of pre-clinical studies.
Collapse
Affiliation(s)
- Miriam Öttl
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081HV, the Netherlands
| | - Ruud F Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081HV, the Netherlands
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081HV, the Netherlands
- Department of Human Genetics, Center for Neurogenomics and Cognitive Research, University Medical Center, De Boelelaan 1117, Amsterdam 1081HV, the Netherlands
| |
Collapse
|
4
|
Gengyo-Ando K, Kumagai M, Ando H, Nakai J. Domain 3a mutation of VPS33A suppresses larval arrest phenotype in the loss of VPS45 in Caenorhabditis elegans. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001155. [PMID: 38585203 PMCID: PMC10995724 DOI: 10.17912/micropub.biology.001155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/09/2024]
Abstract
The Sec1/Munc18 (SM) protein VPS45 is a key regulator of SNARE-mediated membrane fusion in endosomal trafficking, but its precise role remains unknown. To understand the function of VPS45 in vivo , we performed a genetic suppressor screen in Caenorhabditis elegans . We found that the temperature-sensitive lethality caused by the loss of VPS-45 can be suppressed by a mutation in another SM protein, VPS33A. The VPS33A M376I mutation is located in domain 3a, which is predicted to be essential for SNARE complex assembly. These results highlight the functional importance of domain 3a in endosomal SM proteins and its role in specific membrane fusion.
Collapse
Affiliation(s)
- Keiko Gengyo-Ando
- Oral Physiology, Tohoku University Graduate School of Dentistry, Miyagi, Japan
| | - Masahiko Kumagai
- Bioinformatics Unit, Research Center for Advanced Analysis, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Hideki Ando
- Oral Physiology, Tohoku University Graduate School of Dentistry, Miyagi, Japan
| | - Junichi Nakai
- Oral Physiology, Tohoku University Graduate School of Dentistry, Miyagi, Japan
| |
Collapse
|
5
|
Weeratunga S, Gormal RS, Liu M, Eldershaw D, Livingstone EK, Malapaka A, Wallis TP, Bademosi AT, Jiang A, Healy MD, Meunier FA, Collins BM. Interrogation and validation of the interactome of neuronal Munc18-interacting Mint proteins with AlphaFold2. J Biol Chem 2024; 300:105541. [PMID: 38072052 PMCID: PMC10820826 DOI: 10.1016/j.jbc.2023.105541] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 01/13/2024] Open
Abstract
Munc18-interacting proteins (Mints) are multidomain adaptors that regulate neuronal membrane trafficking, signaling, and neurotransmission. Mint1 and Mint2 are highly expressed in the brain with overlapping roles in the regulation of synaptic vesicle fusion required for neurotransmitter release by interacting with the essential synaptic protein Munc18-1. Here, we have used AlphaFold2 to identify and then validate the mechanisms that underpin both the specific interactions of neuronal Mint proteins with Munc18-1 as well as their wider interactome. We found that a short acidic α-helical motif within Mint1 and Mint2 is necessary and sufficient for specific binding to Munc18-1 and binds a conserved surface on Munc18-1 domain3b. In Munc18-1/2 double knockout neurosecretory cells, mutation of the Mint-binding site reduces the ability of Munc18-1 to rescue exocytosis, and although Munc18-1 can interact with Mint and Sx1a (Syntaxin1a) proteins simultaneously in vitro, we find that they have mutually reduced affinities, suggesting an allosteric coupling between the proteins. Using AlphaFold2 to then examine the entire cellular network of putative Mint interactors provides a structural model for their assembly with a variety of known and novel regulatory and cargo proteins including ADP-ribosylation factor (ARF3/ARF4) small GTPases and the AP3 clathrin adaptor complex. Validation of Mint1 interaction with a new predicted binder TJAP1 (tight junction-associated protein 1) provides experimental support that AlphaFold2 can correctly predict interactions across such large-scale datasets. Overall, our data provide insights into the diversity of interactions mediated by the Mint family and show that Mints may help facilitate a key trigger point in SNARE (soluble N-ethylmaleimide-sensitive factor attachment receptor) complex assembly and vesicle fusion.
Collapse
Affiliation(s)
- Saroja Weeratunga
- Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia
| | - Rachel S Gormal
- Clem Jones Centre for Ageing and Dementia Research, Queensland Brain Institute, The University of Queensland, Queensland, Australia
| | - Meihan Liu
- Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia
| | - Denaye Eldershaw
- Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia
| | - Emma K Livingstone
- Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia
| | - Anusha Malapaka
- Clem Jones Centre for Ageing and Dementia Research, Queensland Brain Institute, The University of Queensland, Queensland, Australia
| | - Tristan P Wallis
- Clem Jones Centre for Ageing and Dementia Research, Queensland Brain Institute, The University of Queensland, Queensland, Australia
| | - Adekunle T Bademosi
- Clem Jones Centre for Ageing and Dementia Research, Queensland Brain Institute, The University of Queensland, Queensland, Australia
| | - Anmin Jiang
- Clem Jones Centre for Ageing and Dementia Research, Queensland Brain Institute, The University of Queensland, Queensland, Australia
| | - Michael D Healy
- Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia
| | - Frederic A Meunier
- Clem Jones Centre for Ageing and Dementia Research, Queensland Brain Institute, The University of Queensland, Queensland, Australia; School of Biomedical Sciences, The University of Queensland, Queensland, Australia
| | - Brett M Collins
- Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia.
| |
Collapse
|
6
|
Stefani I, Iwaszkiewicz J, Fasshauer D. Exploring the conformational changes of the Munc18-1/syntaxin 1a complex. Protein Sci 2023; 33:e4870. [PMID: 38109275 PMCID: PMC10895456 DOI: 10.1002/pro.4870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 11/30/2023] [Accepted: 12/07/2023] [Indexed: 12/20/2023]
Abstract
Neurotransmitters are released from synaptic vesicles, the membrane of which fuses with the plasma membrane upon calcium influx. This membrane fusion reaction is driven by the formation of a tight complex comprising the plasma membrane N-ethylmaleimide-sensitive factor attachment receptor (SNARE) proteins syntaxin-1a and SNAP-25 with the vesicle SNARE protein synaptobrevin. The neuronal protein Munc18-1 forms a stable complex with syntaxin-1a. Biochemically, syntaxin-1a cannot escape the tight grip of Munc18-1, so formation of the SNARE complex is inhibited. However, Munc18-1 is essential for the release of neurotransmitters in vivo. It has therefore been assumed that Munc18-1 makes the bound syntaxin-1a available for SNARE complex formation. Exactly how this occurs is still unclear, but it is assumed that structural rearrangements occur. Here, we used a series of mutations to specifically weaken the complex at different positions in order to induce these rearrangements biochemically. Our approach was guided through sequence and structural analysis and supported by molecular dynamics simulations. Subsequently, we created a homology model showing the complex in an altered conformation. This conformation presumably represents a more open arrangement of syntaxin-1a that permits the formation of a SNARE complex to be initiated while still bound to Munc18-1. In the future, research should investigate how this central reaction for neuronal communication is controlled by other proteins.
Collapse
Affiliation(s)
- Ioanna Stefani
- Department of Computational BiologyUniversity of LausanneLausanneSwitzerland
| | | | - Dirk Fasshauer
- Department of Computational BiologyUniversity of LausanneLausanneSwitzerland
| |
Collapse
|
7
|
Wang X, Gong J, Zhu L, Chen H, Jin Z, Mo X, Wang S, Yang X, Ma C. Identification of residues critical for the extension of Munc18-1 domain 3a. BMC Biol 2023; 21:158. [PMID: 37443000 PMCID: PMC10347870 DOI: 10.1186/s12915-023-01655-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND Neurotransmitter release depends on the fusion of synaptic vesicles with the presynaptic membrane and is mainly mediated by SNARE complex assembly. During the transition of Munc18-1/Syntaxin-1 to the SNARE complex, the opening of the Syntaxin-1 linker region catalyzed by Munc13-1 leads to the extension of the domain 3a hinge loop, which enables domain 3a to bind SNARE motifs in Synaptobrevin-2 and Syntaxin-1 and template the SNARE complex assembly. However, the exact mechanism of domain 3a extension remains elusive. RESULTS Here, we characterized residues on the domain 3a hinge loop that are crucial for the extension of domain 3a by using biophysical and biochemical approaches and electrophysiological recordings. We showed that the mutation of residues T323/M324/R325 disrupted Munc13-1-mediated SNARE complex assembly and membrane fusion starting from Munc18-1/Syntaxin-1 in vitro and caused severe defects in the synaptic exocytosis of mouse cortex neurons in vivo. Moreover, the mutation had no effect on the binding of Synaptobrevin-2 to isolated Munc18-1 or the conformational change of the Syntaxin-1 linker region catalyzed by the Munc13-1 MUN domain. However, the extension of the domain 3a hinge loop in Munc18-1/Syntaxin-1 was completely disrupted by the mutation, leading to the failure of Synaptobrevin-2 binding to Munc18-1/Syntaxin-1. CONCLUSIONS Together with previous results, our data further support the model that the template function of Munc18-1 in SNARE complex assembly requires the extension of domain 3a, and particular residues in the domain 3a hinge loop are crucial for the autoinhibitory release of domain 3a after the MUN domain opens the Syntaxin-1 linker region.
Collapse
Affiliation(s)
- Xianping Wang
- Hubei Key Laboratory of Edible Wild Plants Conservation and Utilization, College of Life Sciences, Hubei Normal University, Huangshi, China
| | - Jihong Gong
- Key Laboratory of Cognitive Science, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis & Treatment, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central Minzu University, Wuhan, China
| | - Le Zhu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Huidan Chen
- Key Laboratory of Cognitive Science, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis & Treatment, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central Minzu University, Wuhan, China
| | - Ziqi Jin
- Key Laboratory of Cognitive Science, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis & Treatment, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central Minzu University, Wuhan, China
| | - Xiaoqiang Mo
- Youjiang Medical University for Nationalities, Baise, China
| | - Shen Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofei Yang
- Key Laboratory of Cognitive Science, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis & Treatment, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central Minzu University, Wuhan, China
| | - Cong Ma
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Palfreyman MT, West SE, Jorgensen EM. SNARE Proteins in Synaptic Vesicle Fusion. ADVANCES IN NEUROBIOLOGY 2023; 33:63-118. [PMID: 37615864 DOI: 10.1007/978-3-031-34229-5_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Neurotransmitters are stored in small membrane-bound vesicles at synapses; a subset of synaptic vesicles is docked at release sites. Fusion of docked vesicles with the plasma membrane releases neurotransmitters. Membrane fusion at synapses, as well as all trafficking steps of the secretory pathway, is mediated by SNARE proteins. The SNAREs are the minimal fusion machinery. They zipper from N-termini to membrane-anchored C-termini to form a 4-helix bundle that forces the apposed membranes to fuse. At synapses, the SNAREs comprise a single helix from syntaxin and synaptobrevin; SNAP-25 contributes the other two helices to complete the bundle. Unc13 mediates synaptic vesicle docking and converts syntaxin into the permissive "open" configuration. The SM protein, Unc18, is required to initiate and proofread SNARE assembly. The SNAREs are then held in a half-zippered state by synaptotagmin and complexin. Calcium removes the synaptotagmin and complexin block, and the SNAREs drive vesicle fusion. After fusion, NSF and alpha-SNAP unwind the SNAREs and thereby recharge the system for further rounds of fusion. In this chapter, we will describe the discovery of the SNAREs, their relevant structural features, models for their function, and the central role of Unc18. In addition, we will touch upon the regulation of SNARE complex formation by Unc13, complexin, and synaptotagmin.
Collapse
Affiliation(s)
- Mark T Palfreyman
- School of Biological Sciences, and Howard Hughes Medical Institute, University of Utah, Salt Lake City, UT, USA
| | - Sam E West
- School of Biological Sciences, and Howard Hughes Medical Institute, University of Utah, Salt Lake City, UT, USA
| | - Erik M Jorgensen
- School of Biological Sciences, and Howard Hughes Medical Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
9
|
Huang H, Ouyang Q, Mei K, Liu T, Sun Q, Liu W, Liu R. Acetylation of SCFD1 regulates SNARE complex formation and autophagosome-lysosome fusion. Autophagy 2023; 19:189-203. [PMID: 35465820 PMCID: PMC9809933 DOI: 10.1080/15548627.2022.2064624] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
SCFD1 (sec1 family domain containing 1) was recently shown to function in autophagosome-lysosome fusion, and multiple studies have demonstrated the regulatory impacts of acetylation (a post-translational modification) on macroautophagy/autophagy. Here, we demonstrate that both acetylation- and phosphorylation-dependent mechanisms control SCFD1's function in autophagosome-lysosome fusion. After detecting a decrease in the extent of SCFD1 acetylation under autophagy-stimulated conditions, we found that KAT2B/PCAF catalyzes the acetylation of residues K126 and K515 of SCFD1; we also showed that these two residues are deacetylated by SIRT4. Importantly, we showed that AMPK-controlled SCFD1 phosphorylation strongly disrupts the capacity of SCFD1 to interact with KAT2B, thus ensuring that the SCFD1 acetylation level remains low. Finally, we demonstrated that SCFD1 acetylation inhibits autophagic flux, specifically by blocking STX17-SNAP29-VAMP8 SNARE complex formation. Thus, our study reveals a mechanism through which phosphorylation and acetylation modifications of SCFD1 mediate SNARE complex formation to regulate autophagosome maturation.ACLY: ATP citrate lyase; CREB: cAMP responsive element binding protein; EBSS: nutrient-deprivation medium; EP300: E1A binding protein p300; KAT5/TIP60: lysine acetyltransferase 5; HOPS: homotypic fusion and protein sorting; MS: mass spectroscopy; SCFD1: sec1 family domain containing 1; SM: Sec1/Munc18; SNARE: soluble N-ethylmaleimide-sensitive factor attachment protein receptor; UVRAG: UV radiation resistance associated.
Collapse
Affiliation(s)
- Hong Huang
- College of Food Science and Technology, School of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Qinqin Ouyang
- College of Food Science and Technology, School of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Kunrong Mei
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjing, Tianjing, China
| | - Ting Liu
- Department of Cell Biology, and Department of General Surgery of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qiming Sun
- Department of Biochemistry, and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wei Liu
- Department of Biochemistry, and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China,Zhejiang University School of Medicine, Joint Institute of Genetics and Genomics Medicine between Zhejiang University and University of Toronto, Hangzhou, Zhejiang, China
| | - Rong Liu
- College of Food Science and Technology, School of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, China,Zhejiang University School of Medicine, Lead Contact, Nanjing, China,CONTACT Rong Liu College of Food Science and Technology, School of Life Sciences, Nanjing Agricultural University, Nanjing210095, China
| |
Collapse
|
10
|
Meunier FA, Hu Z. Functional Roles of UNC-13/Munc13 and UNC-18/Munc18 in Neurotransmission. ADVANCES IN NEUROBIOLOGY 2023; 33:203-231. [PMID: 37615868 DOI: 10.1007/978-3-031-34229-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Neurotransmitters are released from synaptic and secretory vesicles following calcium-triggered fusion with the plasma membrane. These exocytotic events are driven by assembly of a ternary SNARE complex between the vesicle SNARE synaptobrevin and the plasma membrane-associated SNAREs syntaxin and SNAP-25. Proteins that affect SNARE complex assembly are therefore important regulators of synaptic strength. In this chapter, we review our current understanding of the roles played by two SNARE interacting proteins: UNC-13/Munc13 and UNC-18/Munc18. We discuss results from both invertebrate and vertebrate model systems, highlighting recent advances, focusing on the current consensus on molecular mechanisms of action and nanoscale organization, and pointing out some unresolved aspects of their functions.
Collapse
Affiliation(s)
- Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia.
| | - Zhitao Hu
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
11
|
Liu F, He R, Zhu M, Zhou L, Liu Y, Yu H. Assembly-promoting protein Munc18c stimulates SNARE-dependent membrane fusion through its SNARE-like peptide. J Biol Chem 2022; 298:102470. [PMID: 36087838 PMCID: PMC9547204 DOI: 10.1016/j.jbc.2022.102470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 11/19/2022] Open
Abstract
Intracellular vesicle fusion requires the soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) and their cognate Sec1/Munc18 (SM) proteins. How SM proteins act in concert with trans-SNARE complexes to promote membrane fusion remains incompletely understood. Munc18c, a broadly distributed SM protein, selectively regulates multiple exocytotic pathways, including GLUT4 exocytosis. Here, using an in vitro reconstituted system, we discovered a SNARE-like peptide (SLP), conserved in Munc18-1 of synaptic exocytosis, is crucial to the stimulatory activity of Munc18c in vesicle fusion. The direct stimulation of the SNARE-mediated fusion reaction by SLP further supported the essential role of this fragment. Interestingly, we found SLP strongly accelerates the membrane fusion rate when anchored to the target membrane but not the vesicle membrane, suggesting it primarily interacts with t-SNAREs in cis to drive fusion. Furthermore, we determined the SLP fragment is competitive with the full-length Munc18c protein and specific to the cognate v-SNARE isoforms, supporting how it could resemble Munc18c’s activity in membrane fusion. Together, our findings demonstrate that Munc18c facilitates SNARE-dependent membrane fusion through SLP, revealing that the t-SNARE-SLP binding mode might be a conserved mechanism for the stimulatory function of SM proteins in vesicle fusion.
Collapse
Affiliation(s)
- Furong Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Ruyue He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Min Zhu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Lin Zhou
- School of Chemistry and Bioengineering, Nanjing Normal University Taizhou College, Taizhou, China
| | - Yinghui Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China.
| | - Haijia Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China.
| |
Collapse
|
12
|
Zhang Y, Ma L, Bao H. Energetics, kinetics, and pathways of SNARE assembly in membrane fusion. Crit Rev Biochem Mol Biol 2022; 57:443-460. [PMID: 36151854 PMCID: PMC9588726 DOI: 10.1080/10409238.2022.2121804] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Fusion of transmitter-containing vesicles with plasma membranes at the synaptic and neuromuscular junctions mediates neurotransmission and muscle contractions, respectively, thereby underlying all thoughts and actions. The fusion process is driven by the coupled folding and assembly of three synaptic SNARE proteins--syntaxin-1 and SNAP-25 on the target plasma membrane (t-SNAREs) and VAMP2 on the vesicular membrane (v-SNARE) into a four-helix bundle. Their assembly is chaperoned by Munc18-1 and many other proteins to achieve the speed and accuracy required for neurotransmission. However, the physiological pathway of SNARE assembly and its coupling to membrane fusion remains unclear. Here, we review recent progress in understanding SNARE assembly and membrane fusion, with a focus on results obtained by single-molecule manipulation approaches and electric recordings of single fusion pores. We describe two pathways of synaptic SNARE assembly, their associated intermediates, energetics, and kinetics. Assembly of the three SNAREs in vitro begins with the formation of a t-SNARE binary complex, on which VAMP2 folds in a stepwise zipper-like fashion. Munc18-1 significantly alters the SNARE assembly pathway: syntaxin-1 and VAMP2 first bind on the surface of Munc18-1 to form a template complex, with which SNAP-25 associates to conclude SNARE assembly and displace Munc18-1. During membrane fusion, multiple trans-SNARE complexes cooperate to open a dynamic fusion pore in a manner dependent upon their copy number and zippering states. Together, these results demonstrate that stepwise and cooperative SNARE assembly drive stagewise membrane fusion.
Collapse
Affiliation(s)
- Yongli Zhang
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA;,Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA;,Conatct: and
| | - Lu Ma
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA;,Present address: Beijing National Laboratory for Condensed Matter Physics and CAS Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
| | - Huan Bao
- Department of Molecular Medicine, The Scripps Research Institute, 130 Scripps Way, Jupiter, Florida, 33458,Conatct: and
| |
Collapse
|
13
|
Parra-Rivas LA, Palfreyman MT, Vu TN, Jorgensen EM. Interspecies complementation identifies a pathway to assemble SNAREs. iScience 2022; 25:104506. [PMID: 35754735 PMCID: PMC9213704 DOI: 10.1016/j.isci.2022.104506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/23/2022] [Accepted: 05/27/2022] [Indexed: 11/18/2022] Open
Abstract
Unc18 and SNARE proteins form the core of the membrane fusion complex at synapses. To understand the functional interactions within the core machinery, we adopted an "interspecies complementation" approach in Caenorhabditis elegans. Substitutions of individual SNAREs and Unc18 proteins with those from yeast fail to rescue fusion. However, synaptic transmission could be restored in worm-yeast chimeras when two key interfaces were present: an Habc-Unc18 contact site and an Unc18-SNARE motif contact site. A constitutively open form of Unc18 bypasses the requirement for the Habc-Unc18 interface. These data suggest that the Habc domain of syntaxin is required for Unc18 to adopt an open conformation; open Unc18 then templates SNARE complex formation. Finally, we demonstrate that the SNARE and Unc18 machinery in the nematode C. elegans can be replaced by yeast proteins and still carry out synaptic transmission, pointing to the deep evolutionary conservation of these two interfaces.
Collapse
Affiliation(s)
- Leonardo A. Parra-Rivas
- Howard Hughes Medical Institute, School of Biological Sciences, University of Utah, Salt Lake City, UT 84112-0840, USA
| | - Mark T. Palfreyman
- Howard Hughes Medical Institute, School of Biological Sciences, University of Utah, Salt Lake City, UT 84112-0840, USA
| | - Thien N. Vu
- Howard Hughes Medical Institute, School of Biological Sciences, University of Utah, Salt Lake City, UT 84112-0840, USA
| | - Erik M. Jorgensen
- Howard Hughes Medical Institute, School of Biological Sciences, University of Utah, Salt Lake City, UT 84112-0840, USA
| |
Collapse
|
14
|
Hwang J, Thurmond DC. Exocytosis Proteins: Typical and Atypical Mechanisms of Action in Skeletal Muscle. Front Endocrinol (Lausanne) 2022; 13:915509. [PMID: 35774142 PMCID: PMC9238359 DOI: 10.3389/fendo.2022.915509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/11/2022] [Indexed: 11/18/2022] Open
Abstract
Insulin-stimulated glucose uptake in skeletal muscle is of fundamental importance to prevent postprandial hyperglycemia, and long-term deficits in insulin-stimulated glucose uptake underlie insulin resistance and type 2 diabetes. Skeletal muscle is responsible for ~80% of the peripheral glucose uptake from circulation via the insulin-responsive glucose transporter GLUT4. GLUT4 is mainly sequestered in intracellular GLUT4 storage vesicles in the basal state. In response to insulin, the GLUT4 storage vesicles rapidly translocate to the plasma membrane, where they undergo vesicle docking, priming, and fusion via the high-affinity interactions among the soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) exocytosis proteins and their regulators. Numerous studies have elucidated that GLUT4 translocation is defective in insulin resistance and type 2 diabetes. Emerging evidence also links defects in several SNAREs and SNARE regulatory proteins to insulin resistance and type 2 diabetes in rodents and humans. Therefore, we highlight the latest research on the role of SNAREs and their regulatory proteins in insulin-stimulated GLUT4 translocation in skeletal muscle. Subsequently, we discuss the novel emerging role of SNARE proteins as interaction partners in pathways not typically thought to involve SNAREs and how these atypical functions reveal novel therapeutic targets for combating peripheral insulin resistance and diabetes.
Collapse
Affiliation(s)
| | - Debbie C. Thurmond
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| |
Collapse
|
15
|
Wang S, Ma C. Neuronal SNARE complex assembly guided by Munc18-1 and Munc13-1. FEBS Open Bio 2022; 12:1939-1957. [PMID: 35278279 PMCID: PMC9623535 DOI: 10.1002/2211-5463.13394] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/28/2022] [Accepted: 03/10/2022] [Indexed: 01/25/2023] Open
Abstract
Neurotransmitter release by Ca2+ -triggered synaptic vesicle exocytosis is essential for information transmission in the nervous system. The soluble N-ethylmaleimide sensitive factor attachment protein receptors (SNAREs) syntaxin-1, SNAP-25, and synaptobrevin-2 form the SNARE complex to bring synaptic vesicles and the plasma membranes together and to catalyze membrane fusion. Munc18-1 and Munc13-1 regulate synaptic vesicle priming via orchestrating neuronal SNARE complex assembly. In this review, we summarize recent advances toward the functions and molecular mechanisms of Munc18-1 and Munc13-1 in guiding neuronal SNARE complex assembly, and discuss the functional similarities and differences between Munc18-1 and Munc13-1 in neurons and their homologs in other intracellular membrane trafficking systems.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Molecular Biophysics of the Ministry of EducationCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Cong Ma
- Key Laboratory of Molecular Biophysics of the Ministry of EducationCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
16
|
Hess JL, Radonjić NV, Patak J, Glatt SJ, Faraone SV. Autophagy, apoptosis, and neurodevelopmental genes might underlie selective brain region vulnerability in attention-deficit/hyperactivity disorder. Mol Psychiatry 2021; 26:6643-6654. [PMID: 33339955 PMCID: PMC8760041 DOI: 10.1038/s41380-020-00974-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Large-scale brain imaging studies by the ENIGMA Consortium identified structural changes associated with attention-deficit/hyperactivity disorder (ADHD). It is not clear why some brain regions are impaired and others spared by the etiological risks for ADHD. We hypothesized that spatial variation in brain cell organization and/or pathway expression levels contribute to selective brain region vulnerability (SBRV) in ADHD. In this study, we used the largest available collection of magnetic resonance imaging (MRI) results from the ADHD ENIGMA Consortium (subcortical MRI n = 3242; cortical MRI n = 4180) along with high-resolution postmortem brain microarray data from Allen Brain Atlas (donors n = 6) from 22 brain regions to investigate our SBRV hypothesis. We performed deconvolution of the bulk transcriptomic data to determine abundances of neuronal and nonneuronal cells in the brain. We assessed the relationships between gene-set expression levels, cell abundance, and standardized effect sizes representing regional changes in brain sizes in cases of ADHD. Our analysis yielded significant correlations between apoptosis, autophagy, and neurodevelopment genes with smaller brain sizes in ADHD, along with associations to regional abundances of astrocytes and oligodendrocytes. The lack of enrichment of common genetic risk variants for ADHD within implicated gene sets suggests an environmental etiology to these differences. This work provides novel mechanistic clues about SBRV in ADHD.
Collapse
Affiliation(s)
- Jonathan L Hess
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Nevena V Radonjić
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Jameson Patak
- Department of Neuroscience, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Stephen J Glatt
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Stephen V Faraone
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
17
|
Abstract
SNARE proteins and Sec1/Munc18 (SM) proteins constitute the core molecular engine that drives nearly all intracellular membrane fusion and exocytosis. While SNAREs are known to couple their folding and assembly to membrane fusion, the physiological pathways of SNARE assembly and the mechanistic roles of SM proteins have long been enigmatic. Here, we review recent advances in understanding the SNARE-SM fusion machinery with an emphasis on biochemical and biophysical studies of proteins that mediate synaptic vesicle fusion. We begin by discussing the energetics, pathways, and kinetics of SNARE folding and assembly in vitro. Then, we describe diverse interactions between SM and SNARE proteins and their potential impact on SNARE assembly in vivo. Recent work provides strong support for the idea that SM proteins function as chaperones, their essential role being to enable fast, accurate SNARE assembly. Finally, we review the evidence that SM proteins collaborate with other SNARE chaperones, especially Munc13-1, and briefly discuss some roles of SNARE and SM protein deficiencies in human disease.
Collapse
Affiliation(s)
- Yongli Zhang
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06520, USA;
| | - Frederick M Hughson
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA;
| |
Collapse
|
18
|
Multi-omics Analysis of the Amygdala in a Rat Chronic Unpredictable Mild Stress Model of Depression. Neuroscience 2021; 463:174-183. [PMID: 33836246 DOI: 10.1016/j.neuroscience.2021.03.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 03/21/2021] [Accepted: 03/26/2021] [Indexed: 01/25/2023]
Abstract
Major depressive disorder is a serious and complex mental illness, and multiple brain regions are involved in its pathogenesis. There is increasing evidence that the amygdala is involved in depression; however, the underlying molecular mechanisms remain unclear. In this study, we applied a combination of liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based metabolomic and isobaric tags for relative and absolute quantitation (iTRAQ) proteomic to study changes in the amygdala in a chronic unpredictable mild stress (CUMS) rat model of depression. Differential analysis identified 42 metabolites and 171 proteins that were differentially expressed in the CUMS and control groups. Integrated analyses revealed two major changes in the amygdala of CUMS rats: (1) perturbations in amino acids and carbohydrate metabolism, transport-/catabolism-related proteins activity, and metabolic enzyme activity; (2) abnormal expression of synaptogenesis and oxidative phosphorylation-associated proteins.
Collapse
|
19
|
Xu YF, Chen X, Yang Z, Xiao P, Liu CH, Li KS, Yang XZ, Wang YJ, Zhu ZL, Xu ZG, Zhang S, Wang C, Song YC, Zhao WD, Wang CH, Ji ZL, Zhang ZY, Cui M, Sun JP, Yu X. PTP-MEG2 regulates quantal size and fusion pore opening through two distinct structural bases and substrates. EMBO Rep 2021; 22:e52141. [PMID: 33764618 DOI: 10.15252/embr.202052141] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/26/2021] [Accepted: 02/18/2021] [Indexed: 02/02/2023] Open
Abstract
Tyrosine phosphorylation of secretion machinery proteins is a crucial regulatory mechanism for exocytosis. However, the participation of protein tyrosine phosphatases (PTPs) in different exocytosis stages has not been defined. Here we demonstrate that PTP-MEG2 controls multiple steps of catecholamine secretion. Biochemical and crystallographic analyses reveal key residues that govern the interaction between PTP-MEG2 and its substrate, a peptide containing the phosphorylated NSF-pY83 site, specify PTP-MEG2 substrate selectivity, and modulate the fusion of catecholamine-containing vesicles. Unexpectedly, delineation of PTP-MEG2 mutants along with the NSF binding interface reveals that PTP-MEG2 controls the fusion pore opening through NSF independent mechanisms. Utilizing bioinformatics search and biochemical and electrochemical screening approaches, we uncover that PTP-MEG2 regulates the opening and extension of the fusion pore by dephosphorylating the DYNAMIN2-pY125 and MUNC18-1-pY145 sites. Further structural and biochemical analyses confirmed the interaction of PTP-MEG2 with MUNC18-1-pY145 or DYNAMIN2-pY125 through a distinct structural basis compared with that of the NSF-pY83 site. Our studies thus provide mechanistic insights in complex exocytosis processes.
Collapse
Affiliation(s)
- Yun-Fei Xu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China.,Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Xu Chen
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Medicine, Jinan, China
| | - Zhao Yang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Peng Xiao
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Chun-Hua Liu
- Department of Physiology, Shandong First Medical University, Taian, China
| | - Kang-Shuai Li
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China.,Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Xiao-Zhen Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yi-Jing Wang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Zhong-Liang Zhu
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Zhi-Gang Xu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, China
| | - Sheng Zhang
- Departments of Medicinal Chemistry and Molecular Pharmacology and of Chemistry, Center for Cancer Research, and Institute for Drug Discovery, Purdue University, West Lafayette, IN, USA
| | - Chuan Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - You-Chen Song
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Wei-Dong Zhao
- Department of Developmental Cell Biology, China Medical University, Shenyang, China
| | - Chang-He Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Zhi-Liang Ji
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Zhong-Yin Zhang
- Departments of Medicinal Chemistry and Molecular Pharmacology and of Chemistry, Center for Cancer Research, and Institute for Drug Discovery, Purdue University, West Lafayette, IN, USA
| | - Min Cui
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Medicine, Jinan, China
| | - Jin-Peng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Medicine, Jinan, China
| |
Collapse
|
20
|
André T, Classen J, Brenner P, Betts MJ, Dörr B, Kreye S, Zuidinga B, Meijer M, Russell RB, Verhage M, Söllner TH. The Interaction of Munc18-1 Helix 11 and 12 with the Central Region of the VAMP2 SNARE Motif Is Essential for SNARE Templating and Synaptic Transmission. eNeuro 2020; 7:ENEURO.0278-20.2020. [PMID: 33055194 PMCID: PMC7768276 DOI: 10.1523/eneuro.0278-20.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/13/2020] [Accepted: 08/24/2020] [Indexed: 01/11/2023] Open
Abstract
Sec1/Munc18 proteins play a key role in initiating the assembly of N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes, the molecular fusion machinery. Employing comparative structure modeling, site specific crosslinking by single amino acid substitutions with the photoactivatable unnatural amino acid p-Benzoyl-phenylalanine (Bpa) and reconstituted vesicle docking/fusion assays, we mapped the binding interface between Munc18-1 and the neuronal v-SNARE VAMP2 with single amino acid resolution. Our results show that helices 11 and 12 of domain 3a in Munc18-1 interact with the VAMP2 SNARE motif covering the region from layers -4 to +5. Residue Q301 in helix 11 plays a pivotal role in VAMP2 binding and template complex formation. A VAMP2 binding deficient mutant, Munc18-1 Q301D, does not stimulate lipid mixing in a reconstituted fusion assay. The neuronal SNARE-organizer Munc13-1, which also binds VAMP2, does not bypass the requirement for the Munc18-1·VAMP2 interaction. Importantly, Munc18-1 Q301D expression in Munc18-1 deficient neurons severely reduces synaptic transmission, demonstrating the physiological significance of the Munc18-1·VAMP2 interaction.
Collapse
Affiliation(s)
- Timon André
- Heidelberg University Biochemistry Center, Heidelberg 69120, Germany
| | | | - Philipp Brenner
- Heidelberg University Biochemistry Center, Heidelberg 69120, Germany
| | - Matthew J Betts
- Heidelberg University Biochemistry Center, Heidelberg 69120, Germany
- BioQuant, Heidelberg University, Heidelberg 69120, Germany
| | - Bernhard Dörr
- Heidelberg University Biochemistry Center, Heidelberg 69120, Germany
| | - Susanne Kreye
- Heidelberg University Biochemistry Center, Heidelberg 69120, Germany
| | | | - Marieke Meijer
- Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR) Amsterdam Neuroscience, VU University and University Medical Center Amsterdam (UMCA), Amsterdam 1081HV, The Netherlands
| | - Robert B Russell
- Heidelberg University Biochemistry Center, Heidelberg 69120, Germany
- BioQuant, Heidelberg University, Heidelberg 69120, Germany
| | - Matthijs Verhage
- Department of Functional Genomics
- Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR) Amsterdam Neuroscience, VU University and University Medical Center Amsterdam (UMCA), Amsterdam 1081HV, The Netherlands
| | - Thomas H Söllner
- Heidelberg University Biochemistry Center, Heidelberg 69120, Germany
| |
Collapse
|
21
|
Eisemann TJ, Allen F, Lau K, Shimamura GR, Jeffrey PD, Hughson FM. The Sec1/Munc18 protein Vps45 holds the Qa-SNARE Tlg2 in an open conformation. eLife 2020; 9:e60724. [PMID: 32804076 PMCID: PMC7470827 DOI: 10.7554/elife.60724] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 08/15/2020] [Indexed: 01/17/2023] Open
Abstract
Fusion of intracellular trafficking vesicles is mediated by the assembly of SNARE proteins into membrane-bridging complexes. SNARE-mediated membrane fusion requires Sec1/Munc18-family (SM) proteins, SNARE chaperones that can function as templates to catalyze SNARE complex assembly. Paradoxically, the SM protein Munc18-1 traps the Qa-SNARE protein syntaxin-1 in an autoinhibited closed conformation. Here we present the structure of a second SM-Qa-SNARE complex, Vps45-Tlg2. Strikingly, Vps45 holds Tlg2 in an open conformation, with its SNARE motif disengaged from its Habc domain and its linker region unfolded. The domain 3a helical hairpin of Vps45 is unfurled, exposing the presumptive R-SNARE binding site to allow template complex formation. Although Tlg2 has a pronounced tendency to form homo-tetramers, Vps45 can rescue Tlg2 tetramers into stoichiometric Vps45-Tlg2 complexes. Our findings demonstrate that SM proteins can engage Qa-SNAREs using at least two different modes, one in which the SNARE is closed and one in which it is open.
Collapse
Affiliation(s)
- Travis J Eisemann
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| | - Frederick Allen
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| | - Kelly Lau
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| | | | - Philip D Jeffrey
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| | | |
Collapse
|
22
|
Wang X, Gong J, Zhu L, Wang S, Yang X, Xu Y, Yang X, Ma C. Munc13 activates the Munc18-1/syntaxin-1 complex and enables Munc18-1 to prime SNARE assembly. EMBO J 2020; 39:e103631. [PMID: 32643828 PMCID: PMC7429736 DOI: 10.15252/embj.2019103631] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 05/20/2020] [Accepted: 06/02/2020] [Indexed: 11/09/2022] Open
Abstract
Priming of synaptic vesicles involves Munc13-catalyzed transition of the Munc18-1/syntaxin-1 complex to the SNARE complex in the presence of SNAP-25 and synaptobrevin-2; Munc13 drives opening of syntaxin-1 via the MUN domain while Munc18-1 primes SNARE assembly via domain 3a. However, the underlying mechanism remains unclear. In this study, we have identified a number of residues in domain 3a of Munc18-1 that are crucial for Munc13 and Munc18-1 actions in SNARE complex assembly and synaptic vesicle priming. Our results showed that two residues (Q301/K308) at the side of domain 3a mediate the interaction between the Munc18-1/syntaxin-1 complex and the MUN domain. This interaction enables the MUN domain to drive the opening of syntaxin-1 linker region, thereby leading to the extension of domain 3a and promoting synaptobrevin-2 binding. In addition, we identified two residues (K332/K333) at the bottom of domain 3a that mediate the interaction between Munc18-1 and the SNARE motif of syntaxin-1. This interaction ensures Munc18-1 to persistently associate with syntaxin-1 during the conformational change of syntaxin-1 from closed to open, which reinforces the role of Munc18-1 in templating SNARE assembly. Taken together, our data suggest a mechanism by which Munc13 activates the Munc18-1/syntaxin-1 complex and enables Munc18-1 to prime SNARE assembly.
Collapse
Affiliation(s)
- Xianping Wang
- Key Laboratory of Molecular Biophysics of the Ministry of EducationCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Jihong Gong
- Key Laboratory of Molecular Biophysics of the Ministry of EducationCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Le Zhu
- Key Laboratory of Molecular Biophysics of the Ministry of EducationCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Shen Wang
- Key Laboratory of Molecular Biophysics of the Ministry of EducationCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Xiaoyu Yang
- Key Laboratory of Molecular Biophysics of the Ministry of EducationCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Yuanyuan Xu
- Key Laboratory of Molecular Biophysics of the Ministry of EducationCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Xiaofei Yang
- Key Laboratory of Cognitive ScienceHubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis & TreatmentLaboratory of Membrane Ion Channels and MedicineCollege of Biomedical EngineeringSouth‐Central University for NationalitiesWuhanChina
| | - Cong Ma
- Key Laboratory of Molecular Biophysics of the Ministry of EducationCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
- Institute of Brain ResearchHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
23
|
Vardar G, Gerth F, Schmitt XJ, Rautenstrauch P, Trimbuch T, Schubert J, Lerche H, Rosenmund C, Freund C. Epilepsy-causing STX1B mutations translate altered protein functions into distinct phenotypes in mouse neurons. Brain 2020; 143:2119-2138. [DOI: 10.1093/brain/awaa151] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/05/2020] [Accepted: 03/25/2020] [Indexed: 01/21/2023] Open
Abstract
AbstractSyntaxin 1B (STX1B) is a core component of the N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex that is critical for the exocytosis of synaptic vesicles in the presynapse. SNARE-mediated vesicle fusion is assisted by Munc18-1, which recruits STX1B in the auto-inhibited conformation, while Munc13 catalyses the fast and efficient pairing of helices during SNARE complex formation. Mutations within the STX1B gene are associated with epilepsy. Here we analysed three STX1B mutations by biochemical and electrophysiological means. These three paradigmatic mutations cause epilepsy syndromes of different severity, from benign fever-associated seizures in childhood to severe epileptic encephalopathies. An insertion/deletion (K45/RMCIE, L46M) mutation (STX1BInDel), causing mild epilepsy and located in the early helical Habc domain, leads to an unfolded protein unable to sustain neurotransmission. STX1BG226R, causing epileptic encephalopathies, strongly compromises the interaction with Munc18-1 and reduces expression of both proteins, the size of the readily releasable pool of vesicles, and Ca2+-triggered neurotransmitter release when expressed in STX1-null neurons. The mutation STX1BV216E, also causing epileptic encephalopathies, only slightly diminishes Munc18-1 and Munc13 interactions, but leads to enhanced fusogenicity and increased vesicular release probability, also in STX1-null neurons. Even though the synaptic output remained unchanged in excitatory hippocampal STX1B+/− neurons exogenously expressing STX1B mutants, the manifestation of clear and distinct molecular disease mechanisms by these mutants suggest that certain forms of epilepsies can be conceptualized by assigning mutations to structurally sensitive regions of the STX1B−Munc18-1 interface, translating into distinct neurophysiological phenotypes.
Collapse
Affiliation(s)
- Gülçin Vardar
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Germany
| | - Fabian Gerth
- Laboratory of Protein Biochemistry, Institute for Chemistry and Biochemistry, Freie Universität Berlin, Germany
| | - Xiao Jakob Schmitt
- Laboratory of Protein Biochemistry, Institute for Chemistry and Biochemistry, Freie Universität Berlin, Germany
| | - Pia Rautenstrauch
- Laboratory of Protein Biochemistry, Institute for Chemistry and Biochemistry, Freie Universität Berlin, Germany
| | - Thorsten Trimbuch
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Germany
| | - Julian Schubert
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | | | - Christian Freund
- Laboratory of Protein Biochemistry, Institute for Chemistry and Biochemistry, Freie Universität Berlin, Germany
| |
Collapse
|
24
|
Rathore SS, Liu Y, Yu H, Wan C, Lee M, Yin Q, Stowell MHB, Shen J. Intracellular Vesicle Fusion Requires a Membrane-Destabilizing Peptide Located at the Juxtamembrane Region of the v-SNARE. Cell Rep 2019; 29:4583-4592.e3. [PMID: 31875562 PMCID: PMC6990648 DOI: 10.1016/j.celrep.2019.11.107] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/13/2019] [Accepted: 11/26/2019] [Indexed: 12/11/2022] Open
Abstract
Intracellular vesicle fusion is mediated by soluble N-ethylmaleimide sensitive factor attachment protein receptors (SNAREs) and Sec1/Munc18 (SM) proteins. It is generally accepted that membrane fusion occurs when the vesicle and target membranes are brought into close proximity by SNAREs and SM proteins. In this work, we demonstrate that, for fusion to occur, membrane bilayers must be destabilized by a conserved membrane-embedded motif located at the juxtamembrane region of the vesicle-anchored v-SNARE. Comprised of basic and hydrophobic residues, the juxtamembrane motif perturbs the lipid bilayer structure and promotes SNARE-SM-mediated membrane fusion. The juxtamembrane motif can be functionally substituted with an unrelated membrane-disrupting peptide in the membrane fusion reaction. These findings establish the juxtamembrane motif of the v-SNARE as a membrane-destabilizing peptide. Requirement of membrane-destabilizing peptides is likely a common feature of biological membrane fusion.
Collapse
Affiliation(s)
- Shailendra S Rathore
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, 347 UCB, Boulder, CO 80309, USA
| | - Yinghui Liu
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, 347 UCB, Boulder, CO 80309, USA; Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Haijia Yu
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, 347 UCB, Boulder, CO 80309, USA; Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| | - Chun Wan
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, 347 UCB, Boulder, CO 80309, USA
| | - MyeongSeon Lee
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, 347 UCB, Boulder, CO 80309, USA
| | - Qian Yin
- Department of Biological Sciences and Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Michael H B Stowell
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, 347 UCB, Boulder, CO 80309, USA
| | - Jingshi Shen
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, 347 UCB, Boulder, CO 80309, USA.
| |
Collapse
|
25
|
In vitro fusion of single synaptic and dense core vesicles reproduces key physiological properties. Nat Commun 2019; 10:3904. [PMID: 31467284 PMCID: PMC6715626 DOI: 10.1038/s41467-019-11873-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 08/02/2019] [Indexed: 12/29/2022] Open
Abstract
Regulated exocytosis of synaptic vesicles is substantially faster than of endocrine dense core vesicles despite similar molecular machineries. The reasons for this difference are unknown and could be due to different regulatory proteins, different spatial arrangements, different vesicle sizes, or other factors. To address these questions, we take a reconstitution approach and compare regulated SNARE-mediated fusion of purified synaptic and dense core chromaffin and insulin vesicles using a single vesicle-supported membrane fusion assay. In all cases, Munc18 and complexin are required to restrict fusion in the absence of calcium. Calcium triggers fusion of all docked vesicles. Munc13 (C1C2MUN domain) is required for synaptic and enhanced insulin vesicle fusion, but not for chromaffin vesicles, correlating inversely with the presence of CAPS protein on purified vesicles. Striking disparities in calcium-triggered fusion rates are observed, increasing with curvature with time constants 0.23 s (synaptic vesicles), 3.3 s (chromaffin vesicles), and 9.1 s (insulin vesicles) and correlating with rate differences in cells.
Collapse
|
26
|
Abstract
The past few years have resulted in an increased awareness and recognition of the prevalence and roles of intrinsically disordered proteins and protein regions (IDPs and IDRs, respectively) in synaptic vesicle trafficking and exocytosis and in overall synaptic organization. IDPs and IDRs constitute a class of proteins and protein regions that lack stable tertiary structure, but nevertheless retain biological function. Their significance in processes such as cell signaling is now well accepted, but their pervasiveness and importance in other areas of biology are not as widely appreciated. Here, we review the prevalence and functional roles of IDPs and IDRs associated with the release and recycling of synaptic vesicles at nerve terminals, as well as with the architecture of these terminals. We hope to promote awareness, especially among neuroscientists, of the importance of this class of proteins in these critical pathways and structures. The examples discussed illustrate some of the ways in which the structural flexibility conferred by intrinsic protein disorder can be functionally advantageous in the context of cellular trafficking and synaptic function.
Collapse
Affiliation(s)
- David Snead
- From the Department of Biochemistry, Weill Cornell Medicine, New York, New York 10021
| | - David Eliezer
- From the Department of Biochemistry, Weill Cornell Medicine, New York, New York 10021
| |
Collapse
|
27
|
Wang S, Li Y, Gong J, Ye S, Yang X, Zhang R, Ma C. Munc18 and Munc13 serve as a functional template to orchestrate neuronal SNARE complex assembly. Nat Commun 2019; 10:69. [PMID: 30622273 PMCID: PMC6325239 DOI: 10.1038/s41467-018-08028-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 12/12/2018] [Indexed: 11/30/2022] Open
Abstract
The transition of the Munc18-1/syntaxin-1 complex to the SNARE complex, a key step involved in exocytosis, is regulated by Munc13-1, SNAP-25 and synaptobrevin-2, but the underlying mechanism remains elusive. Here, we identify an interaction between Munc13-1 and the membrane-proximal linker region of synaptobrevin-2, and reveal its essential role in transition and exocytosis. Upon this interaction, Munc13-1 not only recruits synaptobrevin-2-embedded vesicles to the target membrane but also renders the synaptobrevin-2 SNARE motif more accessible to the Munc18-1/syntaxin-1 complex. Afterward, the entry of SNAP-25 leads to a half-zippered SNARE assembly, which eventually dissociates the Munc18-1/syntaxin-1 complex to complete SNARE complex formation. Our data suggest that Munc18-1 and Munc13-1 together serve as a functional template to orchestrate SNARE complex assembly. Synaptic exocytosis depends on formation of the SNARE complex but its assembly mechanism is still under debate. Here, the authors identify an interaction between Munc13-1 and synaptobrevin-2 that is critical for the transition of the Munc18-1/syntaxin-1 complex to the SNARE complex.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, and the Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, 430074, Wuhan, China
| | - Yun Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, and the Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, 430074, Wuhan, China
| | - Jihong Gong
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, and the Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, 430074, Wuhan, China
| | - Sheng Ye
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China
| | - Xiaofei Yang
- Key Laboratory of Cognitive Science, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis and Treatment, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central University for Nationalities, 430074, Wuhan, China
| | - Rongguang Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China.,National Center for Protein Science Shanghai, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Science, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Cong Ma
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, and the Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, 430074, Wuhan, China.
| |
Collapse
|
28
|
Whitten AE, Jarrott RJ, Hu SH, Duff AP, King GJ, Martin JL, Christie MP. Studying Munc18:Syntaxin Interactions Using Small-Angle Scattering. Methods Mol Biol 2019; 1860:115-144. [PMID: 30317501 DOI: 10.1007/978-1-4939-8760-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The interaction between the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein syntaxin (Sx) and regulatory partner Sec/Munc18 (SM) protein is a critical step in vesicle fusion. The exact role played by SM proteins, whether positive or negative, has been the topic of much debate. High-resolution structures of the SM:Sx complex have shown that SM proteins can bind syntaxin in a closed fusion incompetent state. However, in vitro and in vivo experiments also point to a positive regulatory role for SM proteins that is inconsistent with binding syntaxin in a closed conformation. Here we present protocols we used for the expression and purification of the SM proteins Munc18a and Munc18c and syntaxins 1 and 4 along with procedures used for small-angle X-ray and neutron scattering that showed that syntaxins can bind in an open conformation to SM proteins. We also describe methods for chemical cross-linking experiments and detail how this information can be combined with scattering data to obtain low-resolution structural models for SM:Sx protein complexes.
Collapse
Affiliation(s)
- Andrew E Whitten
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia
| | - Russell J Jarrott
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Shu-Hong Hu
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Anthony P Duff
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia
| | - Gordon J King
- Centre for Microscopy and Microanalysis, The University of Queensland, St Lucia, QLD, Australia
| | - Jennifer L Martin
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Michelle P Christie
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
29
|
Jiao J, He M, Port SA, Baker RW, Xu Y, Qu H, Xiong Y, Wang Y, Jin H, Eisemann TJ, Hughson FM, Zhang Y. Munc18-1 catalyzes neuronal SNARE assembly by templating SNARE association. eLife 2018; 7:41771. [PMID: 30540253 PMCID: PMC6320071 DOI: 10.7554/elife.41771] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/11/2018] [Indexed: 01/16/2023] Open
Abstract
Sec1/Munc18-family (SM) proteins are required for SNARE-mediated membrane fusion, but their mechanism(s) of action remain controversial. Using single-molecule force spectroscopy, we found that the SM protein Munc18-1 catalyzes step-wise zippering of three synaptic SNAREs (syntaxin, VAMP2, and SNAP-25) into a four-helix bundle. Catalysis requires formation of an intermediate template complex in which Munc18-1 juxtaposes the N-terminal regions of the SNARE motifs of syntaxin and VAMP2, while keeping their C-terminal regions separated. SNAP-25 binds the templated SNAREs to induce full SNARE zippering. Munc18-1 mutations modulate the stability of the template complex in a manner consistent with their effects on membrane fusion, indicating that chaperoned SNARE assembly is essential for exocytosis. Two other SM proteins, Munc18-3 and Vps33, similarly chaperone SNARE assembly via a template complex, suggesting that SM protein mechanism is conserved.
Collapse
Affiliation(s)
- Junyi Jiao
- Department of Cell Biology, Yale University School of Medicine, New Haven, United States
| | - Mengze He
- Department of Cell Biology, Yale University School of Medicine, New Haven, United States
| | - Sarah A Port
- Department of Molecular Biology, Princeton University, Princeton, United States
| | - Richard W Baker
- Department of Molecular Biology, Princeton University, Princeton, United States.,Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, United States
| | - Yonggang Xu
- Department of Cell Biology, Yale University School of Medicine, New Haven, United States
| | - Hong Qu
- Department of Cell Biology, Yale University School of Medicine, New Haven, United States
| | - Yujian Xiong
- Department of Cell Biology, Yale University School of Medicine, New Haven, United States
| | - Yukun Wang
- Department of Cell Biology, Yale University School of Medicine, New Haven, United States
| | - Huaizhou Jin
- Department of Cell Biology, Yale University School of Medicine, New Haven, United States
| | - Travis J Eisemann
- Department of Molecular Biology, Princeton University, Princeton, United States
| | - Frederick M Hughson
- Department of Molecular Biology, Princeton University, Princeton, United States
| | - Yongli Zhang
- Department of Cell Biology, Yale University School of Medicine, New Haven, United States
| |
Collapse
|
30
|
Guiberson NGL, Pineda A, Abramov D, Kharel P, Carnazza KE, Wragg RT, Dittman JS, Burré J. Mechanism-based rescue of Munc18-1 dysfunction in varied encephalopathies by chemical chaperones. Nat Commun 2018; 9:3986. [PMID: 30266908 PMCID: PMC6162227 DOI: 10.1038/s41467-018-06507-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 09/07/2018] [Indexed: 12/01/2022] Open
Abstract
Heterozygous de novo mutations in the neuronal protein Munc18-1 are linked to epilepsies, intellectual disability, movement disorders, and neurodegeneration. These devastating diseases have a poor prognosis and no known cure, due to lack of understanding of the underlying disease mechanism. To determine how mutations in Munc18-1 cause disease, we use newly generated S. cerevisiae strains, C. elegans models, and conditional Munc18-1 knockout mouse neurons expressing wild-type or mutant Munc18-1, as well as in vitro studies. We find that at least five disease-linked missense mutations of Munc18-1 result in destabilization and aggregation of the mutant protein. Aggregates of mutant Munc18-1 incorporate wild-type Munc18-1, depleting functional Munc18-1 levels beyond hemizygous levels. We demonstrate that the three chemical chaperones 4-phenylbutyrate, sorbitol, and trehalose reverse the deficits caused by mutations in Munc18-1 in vitro and in vivo in multiple models, offering a novel strategy for the treatment of varied encephalopathies. Munc18-1 is an evolutionary conserved gene whose mutations are linked to various neurological diseases in human. In order to better understand the exact nature of the mutations, the authors here utilize several model systems to show mutant Munc18-1 can aggregate and deplete functional pool of Wt protein, and that chemical chaperones can reverse the cellular deficits.
Collapse
Affiliation(s)
- Noah Guy Lewis Guiberson
- Brain and Mind Research Institute & Appel Institute for Alzheimer's Disease Research, Weill Cornell Medicine, New York, NY, 10021, USA
| | - André Pineda
- Brain and Mind Research Institute & Appel Institute for Alzheimer's Disease Research, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Debra Abramov
- Brain and Mind Research Institute & Appel Institute for Alzheimer's Disease Research, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Parinati Kharel
- Brain and Mind Research Institute & Appel Institute for Alzheimer's Disease Research, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Kathryn E Carnazza
- Brain and Mind Research Institute & Appel Institute for Alzheimer's Disease Research, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Rachel T Wragg
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jeremy S Dittman
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jacqueline Burré
- Brain and Mind Research Institute & Appel Institute for Alzheimer's Disease Research, Weill Cornell Medicine, New York, NY, 10021, USA.
| |
Collapse
|
31
|
Yin P, Gandasi NR, Arora S, Omar-Hmeadi M, Saras J, Barg S. Syntaxin clusters at secretory granules in a munc18-bound conformation. Mol Biol Cell 2018; 29:2700-2708. [PMID: 30156474 PMCID: PMC6249827 DOI: 10.1091/mbc.e17-09-0541] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Syntaxin (stx)-1 is an integral plasma membrane protein that is crucial for two distinct steps of regulated exocytosis, docking of secretory granules at the plasma membrane and membrane fusion. During docking, stx1 clusters at the granule docking site, together with the S/M protein munc18. Here we determined features of stx1 that contribute to its clustering at granules. In live insulin-secreting cells, stx1 and stx3 (but not stx4 or stx11) accumulated at docked granules, and stx1 (but not stx4) rescued docking in cells expressing botulinum neurotoxin-C. Using a series of stx1 deletion mutants and stx1/4 chimeras, we found that all four helical domains (Ha, Hb, Hc, SNARE) and the short N-terminal peptide contribute to recruitment to granules. However, only the Hc domain confers specificity, and it must be derived from stx1 for recruitment to occur. Point mutations in the Hc or the N-terminal peptide designed to interfere with binding to munc18-1 prevent stx1 from clustering at granules, and a mutant munc18 deficient in binding to stx1 does not cluster at granules. We conclude that stx1 is recruited to the docking site in a munc18-1–bound conformation, providing a rationale for the requirement for both proteins for granule docking.
Collapse
Affiliation(s)
- Peng Yin
- Institute of Medical Cell Biology, Uppsala University, 75123 Uppsala, Sweden
| | - Nikhil R Gandasi
- Institute of Medical Cell Biology, Uppsala University, 75123 Uppsala, Sweden
| | - Swati Arora
- Institute of Medical Cell Biology, Uppsala University, 75123 Uppsala, Sweden
| | - Muhmmad Omar-Hmeadi
- Institute of Medical Cell Biology, Uppsala University, 75123 Uppsala, Sweden
| | - Jan Saras
- Institute of Medical Cell Biology, Uppsala University, 75123 Uppsala, Sweden
| | - Sebastian Barg
- Institute of Medical Cell Biology, Uppsala University, 75123 Uppsala, Sweden
| |
Collapse
|
32
|
SNARE zippering requires activation by SNARE-like peptides in Sec1/Munc18 proteins. Proc Natl Acad Sci U S A 2018; 115:E8421-E8429. [PMID: 30127032 DOI: 10.1073/pnas.1802645115] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) catalyze membrane fusion by forming coiled-coil bundles between membrane bilayers. The SNARE bundle zippers progressively toward the membranes, pulling the lipid bilayers into close proximity to fuse. In this work, we found that the +1 and +2 layers in the C-terminal domains (CTDs) of SNAREs are dispensable for reconstituted SNARE-mediated fusion reactions. By contrast, all CTD layers are required for fusion reactions activated by the cognate Sec1/Munc18 (SM) protein or a synthetic Vc peptide derived from the vesicular (v-) SNARE, correlating with strong acceleration of fusion kinetics. These results suggest a similar mechanism underlying the stimulatory functions of SM proteins and Vc peptide in SNARE-dependent membrane fusion. Unexpectedly, we identified a conserved SNARE-like peptide (SLP) in SM proteins that structurally and functionally resembles Vc peptide. Like Vc peptide, SLP binds and activates target (t-) SNAREs, accelerating the fusion reaction. Disruption of the t-SNARE-SLP interaction inhibits exocytosis in vivo. Our findings demonstrated that a t-SNARE-SLP intermediate must form before SNAREs can drive efficient vesicle fusion.
Collapse
|
33
|
Rizo J. Mechanism of neurotransmitter release coming into focus. Protein Sci 2018; 27:1364-1391. [PMID: 29893445 DOI: 10.1002/pro.3445] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 05/10/2018] [Indexed: 12/11/2022]
Abstract
Research for three decades and major recent advances have provided crucial insights into how neurotransmitters are released by Ca2+ -triggered synaptic vesicle exocytosis, leading to reconstitution of basic steps that underlie Ca2+ -dependent membrane fusion and yielding a model that assigns defined functions for central components of the release machinery. The soluble N-ethyl maleimide sensitive factor attachment protein receptors (SNAREs) syntaxin-1, SNAP-25, and synaptobrevin-2 form a tight SNARE complex that brings the vesicle and plasma membranes together and is key for membrane fusion. N-ethyl maleimide sensitive factor (NSF) and soluble NSF attachment proteins (SNAPs) disassemble the SNARE complex to recycle the SNAREs for another round of fusion. Munc18-1 and Munc13-1 orchestrate SNARE complex formation in an NSF-SNAP-resistant manner by a mechanism whereby Munc18-1 binds to synaptobrevin and to a self-inhibited "closed" conformation of syntaxin-1, thus forming a template to assemble the SNARE complex, and Munc13-1 facilitates assembly by bridging the vesicle and plasma membranes and catalyzing opening of syntaxin-1. Synaptotagmin-1 functions as the major Ca2+ sensor that triggers release by binding to membrane phospholipids and to the SNAREs, in a tight interplay with complexins that accelerates membrane fusion. Many of these proteins act as both inhibitors and activators of exocytosis, which is critical for the exquisite regulation of neurotransmitter release. It is still unclear how the actions of these various proteins and multiple other components that control release are integrated and, in particular, how they induce membrane fusion, but it can be expected that these fundamental questions can be answered in the near future, building on the extensive knowledge already available.
Collapse
Affiliation(s)
- Josep Rizo
- Departments of Biophysics, Biochemistry and Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390
| |
Collapse
|
34
|
Abstract
This review summarizes current knowledge of synaptic proteins that are central to synaptic vesicle fusion in presynaptic active zones, including SNAREs (soluble N-ethylmaleimide sensitive factor attachment protein receptors), synaptotagmin, complexin, Munc18 (mammalian uncoordinated-18), and Munc13 (mammalian uncoordinated-13), and highlights recent insights in the cooperation of these proteins for neurotransmitter release. Structural and functional studies of the synaptic fusion machinery suggest new molecular models of synaptic vesicle priming and Ca2+-triggered fusion. These studies will be a stepping-stone toward answering the question of how the synaptic vesicle fusion machinery achieves such high speed and sensitivity.
Collapse
Affiliation(s)
- Axel T Brunger
- Department of Molecular and Cellular Physiology, Department of Neurology and Neurological Sciences, Department of Structural Biology, Department of Photon Science, Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA;
| | - Ucheor B Choi
- Department of Molecular and Cellular Physiology, Department of Neurology and Neurological Sciences, Department of Structural Biology, Department of Photon Science, Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA;
| | - Ying Lai
- Department of Molecular and Cellular Physiology, Department of Neurology and Neurological Sciences, Department of Structural Biology, Department of Photon Science, Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA;
| | - Jeremy Leitz
- Department of Molecular and Cellular Physiology, Department of Neurology and Neurological Sciences, Department of Structural Biology, Department of Photon Science, Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA;
| | - Qiangjun Zhou
- Department of Molecular and Cellular Physiology, Department of Neurology and Neurological Sciences, Department of Structural Biology, Department of Photon Science, Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA;
| |
Collapse
|
35
|
In Vivo Analysis of a Gain-of-Function Mutation Confirms Unc18/Munc18's Role in Priming. J Neurosci 2018; 38:1055-1057. [PMID: 29386300 DOI: 10.1523/jneurosci.3068-17.2017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/19/2017] [Accepted: 12/21/2017] [Indexed: 11/21/2022] Open
|
36
|
Walter AM, Böhme MA, Sigrist SJ. Vesicle release site organization at synaptic active zones. Neurosci Res 2017; 127:3-13. [PMID: 29275162 DOI: 10.1016/j.neures.2017.12.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 12/04/2017] [Accepted: 12/06/2017] [Indexed: 11/30/2022]
Abstract
Information transfer between nerve cells (neurons) forms the basis of behavior, emotion, and survival. Signal transduction from one neuron to another occurs at synapses, and relies on both electrical and chemical signal propagation. At chemical synapses, incoming electrical action potentials trigger the release of chemical neurotransmitters that are sensed by the connected cell and here reconverted to an electrical signal. The presynaptic conversion of an electrical to a chemical signal is an energy demanding, highly regulated process that relies on a complex, evolutionarily conserved molecular machinery. Here, we review the biophysical characteristics of this process, the current knowledge of the molecules operating in this reaction and genetic specializations that may have evolved to shape inter-neuronal signaling.
Collapse
Affiliation(s)
- Alexander M Walter
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Roessle-Straße 10, Berlin 13125, Germany.
| | - Mathias A Böhme
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Roessle-Straße 10, Berlin 13125, Germany
| | - Stephan J Sigrist
- Freie Universität Berlin, Institute for Biology/Genetics, Takustraße 6, 14195 Berlin, Germany; NeuroCure, Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
37
|
Meijer M, Dörr B, Lammertse HC, Blithikioti C, van Weering JR, Toonen RF, Söllner TH, Verhage M. Tyrosine phosphorylation of Munc18-1 inhibits synaptic transmission by preventing SNARE assembly. EMBO J 2017; 37:300-320. [PMID: 29150433 PMCID: PMC5770875 DOI: 10.15252/embj.201796484] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 10/12/2017] [Accepted: 10/16/2017] [Indexed: 11/17/2022] Open
Abstract
Tyrosine kinases are important regulators of synaptic strength. Here, we describe a key component of the synaptic vesicle release machinery, Munc18‐1, as a phosphorylation target for neuronal Src family kinases (SFKs). Phosphomimetic Y473D mutation of a SFK phosphorylation site previously identified by brain phospho‐proteomics abolished the stimulatory effect of Munc18‐1 on SNARE complex formation (“SNARE‐templating”) and membrane fusion in vitro. Furthermore, priming but not docking of synaptic vesicles was disrupted in hippocampal munc18‐1‐null neurons expressing Munc18‐1Y473D. Synaptic transmission was temporarily restored by high‐frequency stimulation, as well as by a Munc18‐1 mutation that results in helix 12 extension, a critical conformational step in vesicle priming. On the other hand, expression of non‐phosphorylatable Munc18‐1 supported normal synaptic transmission. We propose that SFK‐dependent Munc18‐1 phosphorylation may constitute a potent, previously unknown mechanism to shut down synaptic transmission, via direct occlusion of a Synaptobrevin/VAMP2 binding groove and subsequent hindrance of conformational changes in domain 3a responsible for vesicle priming. This would strongly interfere with the essential post‐docking SNARE‐templating role of Munc18‐1, resulting in a largely abolished pool of releasable synaptic vesicles.
Collapse
Affiliation(s)
- Marieke Meijer
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Neuroscience Campus Amsterdam (NCA) VU University Medical Center, Amsterdam, The Netherlands
| | - Bernhard Dörr
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany
| | - Hanna Ca Lammertse
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Neuroscience Campus Amsterdam (NCA) VU University Amsterdam, Amsterdam, The Netherlands
| | - Chrysanthi Blithikioti
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Neuroscience Campus Amsterdam (NCA) VU University Amsterdam, Amsterdam, The Netherlands
| | - Jan Rt van Weering
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Neuroscience Campus Amsterdam (NCA) VU University Medical Center, Amsterdam, The Netherlands
| | - Ruud Fg Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Neuroscience Campus Amsterdam (NCA) VU University Amsterdam, Amsterdam, The Netherlands
| | - Thomas H Söllner
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany
| | - Matthijs Verhage
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Neuroscience Campus Amsterdam (NCA) VU University Medical Center, Amsterdam, The Netherlands .,Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Neuroscience Campus Amsterdam (NCA) VU University Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
38
|
Christie MP, Hu SH, Whitten AE, Rehman A, Jarrott RJ, King GJ, Collins BM, Martin JL. Revisiting interaction specificity reveals neuronal and adipocyte Munc18 membrane fusion regulatory proteins differ in their binding interactions with partner SNARE Syntaxins. PLoS One 2017; 12:e0187302. [PMID: 29088285 PMCID: PMC5663490 DOI: 10.1371/journal.pone.0187302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 10/17/2017] [Indexed: 01/01/2023] Open
Abstract
The efficient delivery of cellular cargo relies on the fusion of cargo-carrying vesicles with the correct membrane at the correct time. These spatiotemporal fusion events occur when SNARE proteins on the vesicle interact with cognate SNARE proteins on the target membrane. Regulatory Munc18 proteins are thought to contribute to SNARE interaction specificity through interaction with the SNARE protein Syntaxin. Neuronal Munc18a interacts with Syntaxin1 but not Syntaxin4, and adipocyte Munc18c interacts with Syntaxin4 but not Syntaxin1. Here we show that this accepted view of specificity needs revision. We find that Munc18c interacts with both Syntaxin4 and Syntaxin1, and appears to bind “non-cognate” Syntaxin1 a little more tightly than Syntaxin4. Munc18a binds Syntaxin1 and Syntaxin4, though it interacts with its cognate Syntaxin1 much more tightly. We also observed that when bound to non-cognate Munc18c, Syntaxin1 captures its neuronal SNARE partners SNAP25 and VAMP2, and Munc18c can bind to pre-formed neuronal SNARE ternary complex. These findings reveal that Munc18a and Munc18c bind Syntaxins differently. Munc18c relies principally on the Syntaxin N-peptide interaction for binding Syntaxin4 or Syntaxin1, whereas Munc18a can bind Syntaxin1 tightly whether or not the Syntaxin1 N-peptide is present. We conclude that Munc18a and Munc18c differ in their binding interactions with Syntaxins: Munc18a has two tight binding modes/sites for Syntaxins as defined previously but Munc18c has just one that requires the N-peptide. These results indicate that the interactions between Munc18 and Syntaxin proteins, and the consequences for in vivo function, are more complex than can be accounted for by binding specificity alone.
Collapse
Affiliation(s)
- Michelle P. Christie
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, University of Queensland, St Lucia, Queensland, Australia
- * E-mail: (MPC); (JLM)
| | - Shu-Hong Hu
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, University of Queensland, St Lucia, Queensland, Australia
| | - Andrew E. Whitten
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, University of Queensland, St Lucia, Queensland, Australia
| | - Asma Rehman
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, University of Queensland, St Lucia, Queensland, Australia
| | - Russell J. Jarrott
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, University of Queensland, St Lucia, Queensland, Australia
| | - Gordon J. King
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, University of Queensland, St Lucia, Queensland, Australia
| | - Brett M. Collins
- Division of Cell Biology and Molecular Medicine, Institute for Molecular Bioscience, University of Queensland, St Lucia, Queensland, Australia
| | - Jennifer L. Martin
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, University of Queensland, St Lucia, Queensland, Australia
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
- * E-mail: (MPC); (JLM)
| |
Collapse
|
39
|
Morey C, Kienle CN, Klöpper TH, Burkhardt P, Fasshauer D. Evidence for a conserved inhibitory binding mode between the membrane fusion assembly factors Munc18 and syntaxin in animals. J Biol Chem 2017; 292:20449-20460. [PMID: 29046354 DOI: 10.1074/jbc.m117.811182] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/10/2017] [Indexed: 12/30/2022] Open
Abstract
The membrane fusion necessary for vesicle trafficking is driven by the assembly of heterologous SNARE proteins orchestrated by the binding of Sec1/Munc18 (SM) proteins to specific syntaxin SNARE proteins. However, the precise mode of interaction between SM proteins and SNAREs is debated, as contrasting binding modes have been found for different members of the SM protein family, including the three vertebrate Munc18 isoforms. While different binding modes could be necessary, given their roles in different secretory processes in different tissues, the structural similarity of the three isoforms makes this divergence perplexing. Although the neuronal isoform Munc18a is well-established to bind tightly to both the closed conformation and the N-peptide of syntaxin 1a, thereby inhibiting SNARE complex formation, Munc18b and -c, which have a more widespread distribution, are reported to mainly interact with the N-peptide of their partnering syntaxins and are thought to instead promote SNARE complex formation. We have reinvestigated the interaction between Munc18c and syntaxin 4 (Syx4). Using isothermal titration calorimetry, we found that Munc18c, like Munc18a, binds to both the closed conformation and the N-peptide of Syx4. Furthermore, using a novel kinetic approach, we found that Munc18c, like Munc18a, slows down SNARE complex formation through high-affinity binding to syntaxin. This strongly suggests that secretory Munc18s in general control the accessibility of the bound syntaxin, probably preparing it for SNARE complex assembly.
Collapse
Affiliation(s)
- Czuee Morey
- From the Département des neurosciences fondamentales, Université de Lausanne, Rue du Bugnon 9, CH-1005 Lausanne, Switzerland
| | - C Nickias Kienle
- From the Département des neurosciences fondamentales, Université de Lausanne, Rue du Bugnon 9, CH-1005 Lausanne, Switzerland
| | - Tobias H Klöpper
- Whitehat Life Sciences Ltd., 20 Wenlock Road, N1 7GU London, United Kingdom, and
| | - Pawel Burkhardt
- the Marine Biological Association, Citadel Hill Marine Laboratory, Plymouth PL1 2PB, United Kingdom
| | - Dirk Fasshauer
- From the Département des neurosciences fondamentales, Université de Lausanne, Rue du Bugnon 9, CH-1005 Lausanne, Switzerland,
| |
Collapse
|
40
|
PKC-dependent phosphorylation of Munc18a at Ser313 in activated RBL-2H3 cells. Inflamm Res 2017; 67:1-3. [PMID: 28983637 DOI: 10.1007/s00011-017-1097-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 09/20/2017] [Accepted: 09/26/2017] [Indexed: 10/18/2022] Open
Abstract
Protein Kinase C (PKC) regulates the release of pro-inflammatory compounds from IgE/antigen-activated mast cells by unknown mechanisms. In this study, we show for the first time that PKC inhibitor Ro-03-0432, which inhibits RBL-2H3 exocytosis/degranulation in a concentration-dependent fashion, prevents the phosphorylation of membrane fusion factor Munc18a at Ser 313. Our study provides fresh evidence that PKC-dependent protein phosphorylation may contribute to the intricate regulation of mast cell degranulation by directly targeting the fusion factors.
Collapse
|
41
|
Munc18a clusters SNARE-bearing liposomes prior to trans-SNARE zippering. Biochem J 2017; 474:3339-3354. [PMID: 28827281 DOI: 10.1042/bcj20170494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/16/2017] [Accepted: 08/17/2017] [Indexed: 12/16/2022]
Abstract
Sec1-Munc18 (SM) proteins co-operate with SNAREs {SNAP [soluble NSF (N-ethylmaleimide-sensitive factor) attachment protein] receptors} to mediate membrane fusion in eukaryotic cells. Studies of Munc18a/Munc18-1/Stxbp1 in neurotransmission suggest that SM proteins accelerate fusion kinetics primarily by activating the partially zippered trans-SNARE complex. However, accumulating evidence has argued for additional roles for SM proteins in earlier steps in the fusion cascade. Here, we investigate the function of Munc18a in reconstituted exocytic reactions mediated by neuronal and non-neuronal SNAREs. We show that Munc18a plays a direct role in promoting proteoliposome clustering, underlying vesicle docking during exocytosis. In the three different fusion reactions examined, Munc18a-dependent clustering requires an intact N-terminal peptide (N-peptide) motif in syntaxin that mediates the binary interaction between syntaxin and Munc18a. Importantly, clustering is preserved under inhibitory conditions that abolish both trans-SNARE complex formation and lipid mixing, indicating that Munc18a promotes membrane clustering in a step that is independent of trans-SNARE zippering and activation.
Collapse
|
42
|
A Calcium- and Diacylglycerol-Stimulated Protein Kinase C (PKC), Caenorhabditis elegans PKC-2, Links Thermal Signals to Learned Behavior by Acting in Sensory Neurons and Intestinal Cells. Mol Cell Biol 2017; 37:MCB.00192-17. [PMID: 28716951 DOI: 10.1128/mcb.00192-17] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/07/2017] [Indexed: 12/18/2022] Open
Abstract
Ca2+- and diacylglycerol (DAG)-activated protein kinase C (cPKC) promotes learning and behavioral plasticity. However, knowledge of in vivo regulation and exact functions of cPKCs that affect behavior is limited. We show that PKC-2, a Caenorhabditis elegans cPKC, is essential for a complex behavior, thermotaxis. C. elegans memorizes a nutrient-associated cultivation temperature (Tc ) and migrates along the Tc within a 17 to 25°C gradient. pkc-2 gene disruption abrogated thermotaxis; a PKC-2 transgene, driven by endogenous pkc-2 promoters, restored thermotaxis behavior in pkc-2-/- animals. Cell-specific manipulation of PKC-2 activity revealed that thermotaxis is controlled by cooperative PKC-2-mediated signaling in both AFD sensory neurons and intestinal cells. Cold-directed migration (cryophilic drive) precedes Tc tracking during thermotaxis. Analysis of temperature-directed behaviors elicited by persistent PKC-2 activation or inhibition in AFD (or intestine) disclosed that PKC-2 regulates initiation and duration of cryophilic drive. In AFD neurons, PKC-2 is a Ca2+ sensor and signal amplifier that operates downstream from cyclic GMP-gated cation channels and distal guanylate cyclases. UNC-18, which regulates neurotransmitter and neuropeptide release from synaptic vesicles, is a critical PKC-2 effector in AFD. UNC-18 variants, created by mutating Ser311 or Ser322, disrupt thermotaxis and suppress PKC-2-dependent cryophilic migration.
Collapse
|
43
|
Rehman A, Hu SH, Tnimov Z, Whitten AE, King GJ, Jarrott RJ, Norwood SJ, Alexandrov K, Collins BM, Christie MP, Martin JL. The nature of the Syntaxin4 C-terminus affects Munc18c-supported SNARE assembly. PLoS One 2017; 12:e0183366. [PMID: 28841669 PMCID: PMC5571939 DOI: 10.1371/journal.pone.0183366] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 08/02/2017] [Indexed: 11/18/2022] Open
Abstract
Vesicular transport of cellular cargo requires targeted membrane fusion and formation of a SNARE protein complex that draws the two apposing fusing membranes together. Insulin-regulated delivery and fusion of glucose transporter-4 storage vesicles at the cell surface is dependent on two key proteins: the SNARE integral membrane protein Syntaxin4 (Sx4) and the soluble regulatory protein Munc18c. Many reported in vitro studies of Munc18c:Sx4 interactions and of SNARE complex formation have used soluble Sx4 constructs lacking the native transmembrane domain. As a consequence, the importance of the Sx4 C-terminal anchor remains poorly understood. Here we show that soluble C-terminally truncated Sx4 dissociates more rapidly from Munc18c than Sx4 where the C-terminal transmembrane domain is replaced with a T4-lysozyme fusion. We also show that Munc18c appears to inhibit SNARE complex formation when soluble C-terminally truncated Sx4 is used but does not inhibit SNARE complex formation when Sx4 is C-terminally anchored (by a C-terminal His-tag bound to resin, by a C-terminal T4L fusion or by the native C-terminal transmembrane domain in detergent micelles). We conclude that the C-terminus of Sx4 is critical for its interaction with Munc18c, and that the reported inhibitory role of Munc18c may be an artifact of experimental design. These results support the notion that a primary role of Munc18c is to support SNARE complex formation and membrane fusion.
Collapse
Affiliation(s)
- Asma Rehman
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Shu-Hong Hu
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Zakir Tnimov
- Division of Cell Biology and Molecular Medicine, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Andrew E. Whitten
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Gordon J. King
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Russell J. Jarrott
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Suzanne J. Norwood
- Division of Cell Biology and Molecular Medicine, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Kirill Alexandrov
- Division of Cell Biology and Molecular Medicine, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Brett M. Collins
- Division of Cell Biology and Molecular Medicine, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Michelle P. Christie
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
- * E-mail: (MPC); (JLM)
| | - Jennifer L. Martin
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
- * E-mail: (MPC); (JLM)
| |
Collapse
|
44
|
UNC-18 and Tomosyn Antagonistically Control Synaptic Vesicle Priming Downstream of UNC-13 in Caenorhabditis elegans. J Neurosci 2017; 37:8797-8815. [PMID: 28821673 DOI: 10.1523/jneurosci.0338-17.2017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 07/21/2017] [Accepted: 08/03/2017] [Indexed: 11/21/2022] Open
Abstract
Munc18-1/UNC-18 is believed to prime SNARE-mediated membrane fusion, yet the underlying mechanisms remain enigmatic. Here, we examine how potential gain-of-function mutations of Munc18-1/UNC-18 affect locomotory behavior and synaptic transmission, and how Munc18-1-mediated priming is related to Munc13-1/UNC-13 and Tomosyn/TOM-1, positive and negative SNARE regulators, respectively. We show that a Munc18-1(P335A)/UNC-18(P334A) mutation leads to significantly increased locomotory activity and acetylcholine release in Caenorhabditis elegans, as well as enhanced synaptic neurotransmission in cultured mammalian neurons. Importantly, similar to tom-1 null mutants, unc-18(P334A) mutants partially bypass the requirement of UNC-13. Moreover, unc-18(P334A) and tom-1 null mutations confer a strong synergy in suppressing the phenotypes of unc-13 mutants. Through biochemical experiments, we demonstrate that Munc18-1(P335A) exhibits enhanced activity in SNARE complex formation as well as in binding to the preformed SNARE complex, and partially bypasses the Munc13-1 requirement in liposome fusion assays. Our results indicate that Munc18-1/UNC-18 primes vesicle fusion downstream of Munc13-1/UNC-13 by templating SNARE complex assembly and acts antagonistically with Tomosyn/TOM-1.SIGNIFICANCE STATEMENT At presynaptic sites, SNARE-mediated membrane fusion is tightly regulated by several key proteins including Munc18/UNC-18, Munc13/UNC-13, and Tomosyn/TOM-1. However, how these proteins interact with each other to achieve the precise regulation of neurotransmitter release remains largely unclear. Using Caenorhabditis elegans as an in vivo model, we found that a gain-of-function mutant of UNC-18 increases locomotory activity and synaptic acetylcholine release, that it partially bypasses the requirement of UNC-13 for release, and that this bypass is synergistically augmented by the lack of TOM-1. We also elucidated the biochemical basis for the gain-of-function caused by this mutation. Thus, our study provides novel mechanistic insights into how Munc18/UNC-18 primes synaptic vesicle release and how this protein interacts functionally with Munc13/UNC-13 and Tomosyn/TOM-1.
Collapse
|
45
|
Extension of Helix 12 in Munc18-1 Induces Vesicle Priming. J Neurosci 2017; 36:6881-91. [PMID: 27358447 DOI: 10.1523/jneurosci.0007-16.2016] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 05/14/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Munc18-1 is essential for vesicle fusion and participates in the docking of large dense-core vesicles to the plasma membrane. Recent structural data suggest that conformational changes in the 12th helix of the Munc18-1 domain 3a within the Munc18-1:syntaxin complex result in an additional interaction with synaptobrevin-2/VAMP2 (vesicle-associated membrane protein 2), leading to SNARE complex formation. To test this hypothesis in living cells, we examined secretion from Munc18-1-null mouse adrenal chromaffin cells expressing Munc18-1 mutants designed to either perturb the extension of helix 12 (Δ324-339), block its interaction with synaptobrevin-2 (L348R), or extend the helix to promote coil-coil interactions with other proteins (P335A). The mutants rescued vesicle docking and syntaxin-1 targeting to the plasma membrane, with the exception of P335A that only supported partial syntaxin-1 targeting. Disruptive mutations (L348R or Δ324-339) lowered the secretory amplitude by decreasing vesicle priming, whereas P335A markedly increased priming and secretory amplitude. The mutants displayed unchanged kinetics and Ca(2+) dependence of fusion, indicating that the mutations specifically affect the vesicle priming step. Mutation of a nearby tyrosine (Y337A), which interacts with closed syntaxin-1, mildly increased secretory amplitude. This correlated with results from an in vitro fusion assay probing the functions of Munc18-1, indicating an easier transition to the extended state in the mutant. Our findings support the notion that a conformational transition within the Munc18-1 domain 3a helix 12 leads to opening of a closed Munc18-1:syntaxin complex, followed by productive SNARE complex assembly and vesicle priming. SIGNIFICANCE STATEMENT The essential postdocking role of Munc18-1 in vesicular exocytosis has remained elusive, but recent data led to the hypothesis that the extension of helix 12 in Munc18 within domain 3a leads to synaptobrevin-2/VAMP2 interaction and SNARE complex formation. Using both lack-of-function and gain-of-function mutants, we here report that the conformation of helix 12 predicts vesicle priming and secretory amplitude in living chromaffin cells. The effects of mutants on secretion could not be explained by differences in syntaxin-1 chaperoning/localization or vesicle docking, and the fusion kinetics and calcium dependence were unchanged, indicating that the effect of helix 12 extension is specific for the vesicle-priming step. We conclude that a conformational change within helix 12 is responsible for the essential postdocking role of Munc18-1 in neurosecretion.
Collapse
|
46
|
Suri M, Evers JMG, Laskowski RA, O'Brien S, Baker K, Clayton-Smith J, Dabir T, Josifova D, Joss S, Kerr B, Kraus A, McEntagart M, Morton J, Smith A, Splitt M, Thornton JM, Wright CF. Protein structure and phenotypic analysis of pathogenic and population missense variants in STXBP1. Mol Genet Genomic Med 2017; 5:495-507. [PMID: 28944233 PMCID: PMC5606886 DOI: 10.1002/mgg3.304] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/17/2017] [Accepted: 05/20/2017] [Indexed: 01/07/2023] Open
Abstract
Background Syntaxin‐binding protein 1, encoded by STXBP1, is highly expressed in the brain and involved in fusing synaptic vesicles with the plasma membrane. Studies have shown that pathogenic loss‐of‐function variants in this gene result in various types of epilepsies, mostly beginning early in life. We were interested to model pathogenic missense variants on the protein structure to investigate the mechanism of pathogenicity and genotype–phenotype correlations. Methods We report 11 patients with pathogenic de novo mutations in STXBP1 identified in the first 4293 trios of the Deciphering Developmental Disorder (DDD) study, including six missense variants. We analyzed the structural locations of the pathogenic missense variants from this study and the literature, as well as population missense variants extracted from Exome Aggregation Consortium (ExAC). Results Pathogenic variants are significantly more likely to occur at highly conserved locations than population variants, and be buried inside the protein domain. Pathogenic mutations are also more likely to destabilize the domain structure compared with population variants, increasing the proportion of (partially) unfolded domains that are prone to aggregation or degradation. We were unable to detect any genotype–phenotype correlation, but unlike previously reported cases, most of the DDD patients with STXBP1 pathogenic variants did not present with very early‐onset or severe epilepsy and encephalopathy, though all have developmental delay with intellectual disability and most display behavioral problems and suffered seizures in later childhood. Conclusion Variants across STXBP1 that cause loss of function can result in severe intellectual disability with or without seizures, consistent with a haploinsufficiency mechanism. Pathogenic missense mutations act through destabilization of the protein domain, making it prone to aggregation or degradation. The presence or absence of early seizures may reflect ascertainment bias in the literature as well as the broad recruitment strategy of the DDD study.
Collapse
Affiliation(s)
- Mohnish Suri
- Nottingham Regional Genetics ServiceNottingham University Hospitals NHS TrustCity Hospital Campus, The Gables, Hucknall RoadNottinghamNG5 1PBUK
| | - Jochem M G Evers
- European Bioinformatics Institute (EMBL-EBI)Wellcome Genome Campus, HinxtonCambridgeCB10 1SDUK
| | - Roman A Laskowski
- European Bioinformatics Institute (EMBL-EBI)Wellcome Genome Campus, HinxtonCambridgeCB10 1SDUK
| | - Sinead O'Brien
- MRC Cognition and Brain Sciences Unit15 Chaucer RoadCambridgeCB2 7EFUK
| | - Kate Baker
- MRC Cognition and Brain Sciences Unit15 Chaucer RoadCambridgeCB2 7EFUK.,Department of Medical GeneticsUniversity of CambridgeCambridge Biomedical CampusCambridgeCB2 0QQUK
| | - Jill Clayton-Smith
- Manchester Centre for Genomic MedicineSt Mary's Hospital, Central Manchester University Hospitals NHS Foundation TrustManchester Academic Health Science CentreManchesterM13 9WLUK
| | - Tabib Dabir
- Northern Ireland Regional Genetics CentreBelfast Health and Social Care TrustBelfast City HospitalLisburn RoadBelfastBT9 7ABUK
| | - Dragana Josifova
- South East Thames Regional Genetics CentreGuy's and St Thomas' NHS Foundation TrustGuy's HospitalGreat Maze PondLondonSE1 9RTUK
| | - Shelagh Joss
- West of Scotland Genetics ServiceQueen Elizabeth University HospitalLaboratory Medicine BuildingGlasgowG51 4TFUK
| | - Bronwyn Kerr
- Manchester Centre for Genomic MedicineSt Mary's Hospital, Central Manchester University Hospitals NHS Foundation TrustManchester Academic Health Science CentreManchesterM13 9WLUK
| | - Alison Kraus
- Yorkshire Regional Genetics ServiceDepartment of Clinical GeneticsLeeds Teaching Hospitals NHS TrustChapel Allerton HospitalChapeltown RoadLeedsLS7 4SAUK
| | - Meriel McEntagart
- South West Thames Regional Genetics CentreSt George's Healthcare NHS TrustSt George's University of LondonCranmer TerraceLondonSW17 0REUK
| | - Jenny Morton
- West Midlands Regional Clinical Genetics Service and Birmingham Health PartnersBirmingham Women's and Children's NHS Foundation TrustBirmingham Women's HospitalMindelsohn Way, EdgbastonBirminghamB15 2TGUK
| | - Audrey Smith
- Yorkshire Regional Genetics ServiceDepartment of Clinical GeneticsLeeds Teaching Hospitals NHS TrustChapel Allerton HospitalChapeltown RoadLeedsLS7 4SAUK
| | - Miranda Splitt
- Northern Genetics ServiceNewcastle upon Tyne Hospitals NHS Foundation TrustInstitute of Human GeneticsInternational Centre for LifeCentral ParkwayNewcastle upon TyneNE1 3BZUK
| | - Janet M Thornton
- European Bioinformatics Institute (EMBL-EBI)Wellcome Genome Campus, HinxtonCambridgeCB10 1SDUK
| | | | - Caroline F Wright
- Wellcome Trust Sanger InstituteWellcome Genome Campus, HinxtonCambridgeCB1 8RQUK.,University of Exeter Medical SchoolRoyal Devon & Exeter HospitalBarrack RoadExeterEX2 5DWUK
| |
Collapse
|
47
|
Sitarska E, Xu J, Park S, Liu X, Quade B, Stepien K, Sugita K, Brautigam CA, Sugita S, Rizo J. Autoinhibition of Munc18-1 modulates synaptobrevin binding and helps to enable Munc13-dependent regulation of membrane fusion. eLife 2017; 6. [PMID: 28477408 PMCID: PMC5464772 DOI: 10.7554/elife.24278] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 05/04/2017] [Indexed: 01/08/2023] Open
Abstract
Munc18-1 orchestrates SNARE complex assembly together with Munc13-1 to mediate neurotransmitter release. Munc18-1 binds to synaptobrevin, but the relevance of this interaction and its relation to Munc13 function are unclear. NMR experiments now show that Munc18-1 binds specifically and non-specifically to synaptobrevin. Specific binding is inhibited by a L348R mutation in Munc18-1 and enhanced by a D326K mutation designed to disrupt the ‘furled conformation’ of a Munc18-1 loop. Correspondingly, the activity of Munc18-1 in reconstitution assays that require Munc18-1 and Munc13-1 for membrane fusion is stimulated by the D326K mutation and inhibited by the L348R mutation. Moreover, the D326K mutation allows Munc13-1-independent fusion and leads to a gain-of-function in rescue experiments in Caenorhabditis elegans unc-18 nulls. Together with previous studies, our data support a model whereby Munc18-1 acts as a template for SNARE complex assembly, and autoinhibition of synaptobrevin binding contributes to enabling regulation of neurotransmitter release by Munc13-1. DOI:http://dx.doi.org/10.7554/eLife.24278.001 Nerve cells communicate with other nerve cells by releasing small molecules called neurotransmitters. The neurotransmitters are first packaged inside bubble-like structures called vesicles, which fuse with the membrane of the nerve cell when it is stimulated. Once the vesicle and membrane have fused, the neurotransmitters are released outside the nerve cell and are detected when they bind to proteins on the surface of other nearby nerve cells. A machinery of different proteins controls membrane fusion. Amongst these proteins are five called Munc18-1, Munc13-1, syntaxin-1, synaptobrevin and SNAP-25. The last three form a tight bundle called SNARE complex that brings the vesicle and cell membrane together and is essential for the two to fuse. Munc18-1 and Munc13-1 orchestrate the assembly of the SNARE complex. Previous studies suggested that Munc18-1 binds to synaptobrevin, providing a template to bring syntaxin-1 and synaptobrevin together and thereby helping the SNARE complex to form. However, the importance of the interaction between Munc18-1 and synaptobrevin was not clearly established, and it was not known how Munc13-1 is involved. Sitarska, Xu et al. have now measured how mutated versions of Munc18-1 bind to synaptobrevin and tested how the mutations affect membrane fusion. A mutation in Munc18-1 that increased binding to synaptobrevin increased membrane fusion too, while a mutation that decreased binding had the opposite effect and reduced fusion. The results support the idea that Munc18-1 provides a template for the SNARE complex to form. One mutation stimulated Munc18-1 so that Munc13-1 was no longer needed for fusion when the mutant Munc18-1 was tested in fusion assays with artificial membranes. This mutation was designed to perturb the structure of a region of Munc18-1 protein that normally inhibits the binding of synaptobrevin. These results suggest that by adopting a state where it cannot bind synaptobrevin, Munc18-1 can only be stimulated to form the SNARE complex and trigger release of neurotransmitter when Munc13-1 is present. This provides a way for Munc13-1, which is regulated by many factors, to fine-tune the release of neurotransmitter. Future work will test whether these proteins work in the same way in living animals. This will help us understand how communication between neurons is finely controlled to enable the brain to carry out its many different tasks. DOI:http://dx.doi.org/10.7554/eLife.24278.002
Collapse
Affiliation(s)
- Ewa Sitarska
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Junjie Xu
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Seungmee Park
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Xiaoxia Liu
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Bradley Quade
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Karolina Stepien
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Kyoko Sugita
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Chad A Brautigam
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Shuzo Sugita
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Josep Rizo
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
48
|
Santos TC, Wierda K, Broeke JH, Toonen RF, Verhage M. Early Golgi Abnormalities and Neurodegeneration upon Loss of Presynaptic Proteins Munc18-1, Syntaxin-1, or SNAP-25. J Neurosci 2017; 37:4525-4539. [PMID: 28348137 PMCID: PMC6596660 DOI: 10.1523/jneurosci.3352-16.2017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 02/07/2017] [Accepted: 03/07/2017] [Indexed: 11/21/2022] Open
Abstract
The loss of presynaptic proteins Munc18-1, syntaxin-1, or SNAP-25 is known to produce cell death, but the underlying features have not been compared experimentally. Here, we investigated these features in cultured mouse CNS and DRG neurons. Side-by-side comparisons confirmed massive cell death, before synaptogenesis, within 1-4 DIV upon loss of t-SNAREs (syntaxin-1, SNAP-25) or Munc18-1, but not v-SNAREs (synaptobrevins/VAMP1/2/3 using tetanus neurotoxin (TeNT), also in TI-VAMP/VAMP7 knock-out (KO) neurons). A condensed cis-Golgi was the first abnormality observed upon Munc18-1 or SNAP-25 loss within 3 DIV. This phenotype was distinct from the Golgi fragmentation observed in apoptosis. Cell death was too rapid after syntaxin-1 loss to study Golgi abnormalities. Syntaxin-1 and Munc18-1 depend on each other for normal cellular levels. We observed that endogenous syntaxin-1 accumulates at the Golgi of Munc18-1 KO neurons. However, expression of a non-neuronal Munc18 isoform that does not bind syntaxin-1, Munc18-3, in Munc18-1 KO neurons prevented cell death and restored normal cis-Golgi morphology, but not synaptic transmission or syntaxin-1 targeting. Finally, we observed that DRG neurons are the only Munc18-1 KO neurons that do not degenerate in vivo or in vitro In these neurons, cis-Golgi abnormalities were less severe, with no changes in Golgi shape. Together, these data demonstrate that cell death upon Munc18-1, syntaxin-1, or SNAP-25 loss occurs via a degenerative pathway unrelated to the known synapse function of these proteins and involving early cis-Golgi abnormalities, distinct from apoptosis.SIGNIFICANCE STATEMENT This study provides new insights in a neurodegeneration pathway triggered by the absence of specific proteins involved in synaptic transmission (syntaxin-1, Munc18-1, SNAP-25), whereas other proteins involved in the same molecular process (synaptobrevins, Munc13-1/2) do not cause degeneration. Massive cell death occurs in cultured neurons upon depleting syntaxin-1, Munc18-1, and/or SNAP-25, well before synapse formation. This study characterizes several relevant cellular phenotypes, especially early cis-Golgi abnormalities, distinct from abnormalities observed during apoptosis, and rules out several other phenotypes as causal (defects in syntaxin-1 targeting and synaptic transmission). As proteins, such as syntaxin-1, Munc18-1, or SNAP-25, modulate α-synuclein neuropathy and/or are dysregulated in Alzheimer's disease, understanding this type of neurodegeneration may provide new links between synaptic defects and neurodegeneration in humans.
Collapse
Affiliation(s)
| | | | - Jurjen H Broeke
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, 1081 HV Amsterdam, The Netherlands
| | | | - Matthijs Verhage
- Department of Functional Genomics and
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam and VU Medical Center, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
49
|
Aslamy A, Thurmond DC. Exocytosis proteins as novel targets for diabetes prevention and/or remediation? Am J Physiol Regul Integr Comp Physiol 2017; 312:R739-R752. [PMID: 28356294 DOI: 10.1152/ajpregu.00002.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/24/2017] [Accepted: 03/24/2017] [Indexed: 12/17/2022]
Abstract
Diabetes remains one of the leading causes of morbidity and mortality worldwide, affecting an estimated 422 million adults. In the US, it is predicted that one in every three children born as of 2000 will suffer from diabetes in their lifetime. Type 2 diabetes results from combinatorial defects in pancreatic β-cell glucose-stimulated insulin secretion and in peripheral glucose uptake. Both processes, insulin secretion and glucose uptake, are mediated by exocytosis proteins, SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) complexes, Sec1/Munc18 (SM), and double C2-domain protein B (DOC2B). Increasing evidence links deficiencies in these exocytosis proteins to diabetes in rodents and humans. Given this, emerging studies aimed at restoring and/or enhancing cellular levels of certain exocytosis proteins point to promising outcomes in maintaining functional β-cell mass and enhancing insulin sensitivity. In doing so, new evidence also shows that enhancing exocytosis protein levels may promote health span and longevity and may also harbor anti-cancer and anti-Alzheimer's disease capabilities. Herein, we present a comprehensive review of the described capabilities of certain exocytosis proteins and how these might be targeted for improving metabolic dysregulation.
Collapse
Affiliation(s)
- Arianne Aslamy
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana; and
| | - Debbie C Thurmond
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana; and .,Department of Molecular and Cellular Endocrinology, Beckman Research Institute of City of Hope, Duarte, California
| |
Collapse
|
50
|
Wang S, Choi UB, Gong J, Yang X, Li Y, Wang AL, Yang X, Brunger AT, Ma C. Conformational change of syntaxin linker region induced by Munc13s initiates SNARE complex formation in synaptic exocytosis. EMBO J 2017; 36:816-829. [PMID: 28137749 PMCID: PMC5350566 DOI: 10.15252/embj.201695775] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 01/01/2017] [Accepted: 01/04/2017] [Indexed: 01/04/2023] Open
Abstract
The soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein syntaxin-1 adopts a closed conformation when bound to Munc18-1, preventing binding to synaptobrevin-2 and SNAP-25 to form the ternary SNARE complex. Although it is known that the MUN domain of Munc13-1 catalyzes the transition from the Munc18-1/syntaxin-1 complex to the SNARE complex, the molecular mechanism is unclear. Here, we identified two conserved residues (R151, I155) in the syntaxin-1 linker region as key sites for the MUN domain interaction. This interaction is essential for SNARE complex formation in vitro and synaptic vesicle priming in neuronal cultures. Moreover, this interaction is important for a tripartite Munc18-1/syntaxin-1/MUN complex, in which syntaxin-1 still adopts a closed conformation tightly bound to Munc18-1, whereas the syntaxin-1 linker region changes its conformation, similar to that of the LE mutant of syntaxin-1 when bound to Munc18-1. We suggest that the conformational change of the syntaxin-1 linker region induced by Munc13-1 initiates ternary SNARE complex formation in the neuronal system.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology and the Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Ucheor B Choi
- Departments of Molecular and Cellular Physiology, Neurology and Neurological Sciences, Photon Science, and Structural Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Jihong Gong
- Key Laboratory of Cognitive Science, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis & Treatment, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, College of Life Science, South-Central University for Nationalities, Wuhan, China
| | - Xiaoyu Yang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology and the Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Li
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology and the Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Austin L Wang
- Departments of Molecular and Cellular Physiology, Neurology and Neurological Sciences, Photon Science, and Structural Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Xiaofei Yang
- Key Laboratory of Cognitive Science, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis & Treatment, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, College of Life Science, South-Central University for Nationalities, Wuhan, China
| | - Axel T Brunger
- Departments of Molecular and Cellular Physiology, Neurology and Neurological Sciences, Photon Science, and Structural Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Cong Ma
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology and the Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|