1
|
Park PY, Bleakley LE, Saraya N, Al-Jawahiri R, Eck J, Aloi MA, Melland H, Baker K, Gordon SL. Correlation between evoked neurotransmitter release and adaptive functions in SYT1-associated neurodevelopmental disorder. EBioMedicine 2024; 109:105416. [PMID: 39481209 DOI: 10.1016/j.ebiom.2024.105416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Pathogenic missense variants in the essential synaptic vesicle protein synaptotagmin-1 (SYT1) cause a neurodevelopmental disorder characterised by motor delay and intellectual disability, hyperkinetic movement disorder, episodic agitation, and visual impairments. SYT1 is the presynaptic calcium sensor that triggers synchronous neurotransmitter release. We have previously shown that pathogenic variants around the calcium-sensing region of the critical C2B domain decrease synaptic vesicle exocytosis in neurons. METHODS Here, we have used cultured hippocampal neurons transfected with SYT1-pHluorin to examine how variants within the C2A and C2B domain of SYT1 impact evoked exocytosis. FINDINGS We show that recently identified variants within the facilitatory C2A domain of the protein (L159R, T196K, E209K, E219Q), as well as additional variants in the C2B domain (M303V, S309P, Y365C, G369D), share an underlying pathogenic mechanism, causing a graded and variant-dependent dominant-negative impairment in exocytosis. We establish that the extent of evoked exocytosis observed in vitro in the presence of SYT1 variants correlates with neurodevelopmental impacts of this disorder. Specifically, the severity of motor and communication impairments exhibited by individuals harbouring these variants correlates with multiple measures of exocytic efficiency. INTERPRETATION Together, this suggests that there is a genotype-function-phenotype relationship in SYT1-associated neurodevelopmental disorder, centring impaired evoked neurotransmitter release as a common pathogenic driver. Moreover, this points toward a direct link between control of neurotransmitter release and development of adaptive functions, providing a tractable target for therapeutic amelioration. FUNDING Australian National Health and Medical Research Council, UK Medical Research Council, Great Ormond Street Hospital Children's Charity, University of Melbourne.
Collapse
Affiliation(s)
- Paul Yangho Park
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, 3052, VIC, Australia
| | - Lauren Elizabeth Bleakley
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, 3052, VIC, Australia
| | - Nadia Saraya
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, 3052, VIC, Australia
| | - Reem Al-Jawahiri
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, CB2 7EF, UK
| | - Josefine Eck
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, CB2 7EF, UK
| | - Marc Anthony Aloi
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, 3052, VIC, Australia
| | - Holly Melland
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, 3052, VIC, Australia; Neuromedicines Discovery Centre, Monash University, Parkville, 3052, VIC, Australia
| | - Kate Baker
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, CB2 7EF, UK; Department of Medical Genetics, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Sarah Louise Gordon
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, 3052, VIC, Australia.
| |
Collapse
|
2
|
Toulmé E, Salazar Lázaro A, Trimbuch T, Rizo J, Rosenmund C. Neurotransmitter release is triggered by a calcium-induced rearrangement in the Synaptotagmin-1/SNARE complex primary interface. Proc Natl Acad Sci U S A 2024; 121:e2409636121. [PMID: 39374398 PMCID: PMC11494337 DOI: 10.1073/pnas.2409636121] [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: 05/14/2024] [Accepted: 08/14/2024] [Indexed: 10/09/2024] Open
Abstract
The Ca2+ sensor synaptotagmin-1 (Syt1) triggers neurotransmitter release together with the neuronal sensitive factor attachment protein receptor (SNARE) complex formed by syntaxin-1, SNAP25, and synaptobrevin. Moreover, Syt1 increases synaptic vesicle (SV) priming and impairs spontaneous vesicle release. The Syt1 C2B domain binds to the SNARE complex through a primary interface via two regions (I and II), but how exactly this interface mediates distinct functions of Syt1 and the mechanism underlying Ca2+ triggering of release are unknown. Using mutagenesis and electrophysiological experiments, we show that region II is functionally and spatially subdivided: Binding of C2B domain arginines to SNAP-25 acidic residues at one face of region II is crucial for Ca2+-evoked release but not for vesicle priming or clamping of spontaneous release, whereas other SNAP-25 and syntaxin-1 acidic residues at the other face mediate priming and clamping of spontaneous release but not evoked release. Mutations that disrupt region I impair the priming and clamping functions of Syt1 while, strikingly, mutations that enhance binding through this region increase vesicle priming and clamping of spontaneous release, but strongly inhibit evoked release and vesicle fusogenicity. These results support previous findings that the primary interface mediates the functions of Syt1 in vesicle priming and clamping of spontaneous release and, importantly, show that Ca2+ triggering of release requires a rearrangement of the primary interface involving dissociation of region I, while region II remains bound. Together with biophysical studies presented in [K. Jaczynska et al., bioRxiv [Preprint] (2024). https://doi.org/10.1101/2024.06.17.599417 (Accessed 18 June 2024)], our data suggest a model whereby this rearrangement pulls the SNARE complex to facilitate fast SV fusion.
Collapse
Affiliation(s)
- Estelle Toulmé
- Institute of Neurophysiology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin10117, Germany
- NeuroCure Cluster of Excellence, Berlin10117, Germany
| | - Andrea Salazar Lázaro
- Institute of Neurophysiology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin10117, Germany
- NeuroCure Cluster of Excellence, Berlin10117, Germany
| | - Thorsten Trimbuch
- Institute of Neurophysiology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin10117, Germany
- NeuroCure Cluster of Excellence, Berlin10117, Germany
| | - Josep Rizo
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Christian Rosenmund
- Institute of Neurophysiology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin10117, Germany
- NeuroCure Cluster of Excellence, Berlin10117, Germany
| |
Collapse
|
3
|
Toulmé E, Lázaro AS, Trimbuch T, Rizo J, Rosenmund C. Neurotransmitter release is triggered by a calcium-induced rearrangement in the Synaptotagmin-1/SNARE complex primary interface. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599435. [PMID: 38948868 PMCID: PMC11213007 DOI: 10.1101/2024.06.17.599435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The Ca2+ sensor synaptotagmin-1 triggers neurotransmitter release together with the neuronal SNARE complex formed by syntaxin-1, SNAP25 and synaptobrevin. Moreover, synaptotagmin-1 increases synaptic vesicle priming and impairs spontaneous vesicle release. The synaptotagmin-1 C2B domain binds to the SNARE complex through a primary interface via two regions (I and II), but how exactly this interface mediates distinct functions of synaptotagmin-1, and the mechanism underlying Ca2+-triggering of release is unknown. Using mutagenesis and electrophysiological experiments, we show that region II is functionally and spatially subdivided: binding of C2B domain arginines to SNAP-25 acidic residues at one face of region II is crucial for Ca2+-evoked release but not for vesicle priming or clamping of spontaneous release, whereas other SNAP-25 and syntaxin-1 acidic residues at the other face mediate priming and clamping of spontaneous release but not evoked release. Mutations that disrupt region I impair the priming and clamping functions of synaptotagmin-1 while, strikingly, mutations that enhance binding through this region increase vesicle priming and clamping of spontaneous release, but strongly inhibit evoked release and vesicle fusogenicity. These results support previous findings that the primary interface mediates the functions of synaptotagmin-1 in vesicle priming and clamping of spontaneous release, and, importantly, show that Ca2+-triggering of release requires a rearrangement of the primary interface involving dissociation of region I, while region II remains bound. Together with modeling and biophysical studies presented in the accompanying paper, our data suggest a model whereby this rearrangement pulls the SNARE complex to facilitate fast synaptic vesicle fusion.
Collapse
Affiliation(s)
- Estelle Toulmé
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Berlin, Germany
- NeuroCure Cluster of Excellence, Berlin, Germany
| | - Andrea Salazar Lázaro
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Berlin, Germany
- NeuroCure Cluster of Excellence, Berlin, Germany
| | - Thorsten Trimbuch
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Berlin, Germany
- NeuroCure Cluster of Excellence, Berlin, Germany
| | - Josep Rizo
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Christian Rosenmund
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Berlin, Germany
- NeuroCure Cluster of Excellence, Berlin, Germany
| |
Collapse
|
4
|
Jaczynska K, Esser V, Xu J, Sari L, Lin MM, Rosenmund C, Rizo J. A lever hypothesis for Synaptotagmin-1 action in neurotransmitter release. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599417. [PMID: 38948826 PMCID: PMC11212951 DOI: 10.1101/2024.06.17.599417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Neurotransmitter release is triggered in microseconds by Ca2+-binding to the Synaptotagmin-1 C2 domains and by SNARE complexes that form four-helix bundles between synaptic vesicles and plasma membranes, but the coupling mechanism between Ca2+-sensing and membrane fusion is unknown. Release requires extension of SNARE helices into juxtamembrane linkers that precede transmembrane regions (linker zippering) and binding of the Synaptotagmin-1 C2B domain to SNARE complexes through a 'primary interface' comprising two regions (I and II). The Synaptotagmin-1 Ca2+-binding loops were believed to accelerate membrane fusion by inducing membrane curvature, perturbing lipid bilayers or helping bridge the membranes, but SNARE complex binding orients the Ca2+-binding loops away from the fusion site, hindering these putative activities. Molecular dynamics simulations now suggest that Synaptotagmin-1 C2 domains near the site of fusion hinder SNARE action, providing an explanation for this paradox and arguing against previous models of Sytnaptotagmin-1 action. NMR experiments reveal that binding of C2B domain arginines to SNARE acidic residues at region II remains after disruption of region I. These results and fluorescence resonance energy transfer assays, together with previous data, suggest that Ca2+ causes reorientation of the C2B domain on the membrane and dissociation from the SNAREs at region I but not region II. Based on these results and molecular modeling, we propose that Synaptotagmin-1 acts as a lever that pulls the SNARE complex when Ca2+ causes reorientation of the C2B domain, facilitating linker zippering and fast membrane fusion. This hypothesis is supported by the electrophysiological data described in the accompanying paper.
Collapse
Affiliation(s)
- Klaudia Jaczynska
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Victoria Esser
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Junjie Xu
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Levent Sari
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Green Center for Systems Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Milo M. Lin
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Green Center for Systems Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Christian Rosenmund
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Berlin, Germany
- NeuroCure Cluster of Excellence, Berlin, Germany
| | - Josep Rizo
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
5
|
Kádková A, Murach J, Østergaard M, Malsam A, Malsam J, Lolicato F, Nickel W, Söllner TH, Sørensen JB. SNAP25 disease mutations change the energy landscape for synaptic exocytosis due to aberrant SNARE interactions. eLife 2024; 12:RP88619. [PMID: 38411501 PMCID: PMC10911398 DOI: 10.7554/elife.88619] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024] Open
Abstract
SNAP25 is one of three neuronal SNAREs driving synaptic vesicle exocytosis. We studied three mutations in SNAP25 that cause epileptic encephalopathy: V48F, and D166Y in the synaptotagmin-1 (Syt1)-binding interface, and I67N, which destabilizes the SNARE complex. All three mutations reduced Syt1-dependent vesicle docking to SNARE-carrying liposomes and Ca2+-stimulated membrane fusion in vitro and when expressed in mouse hippocampal neurons. The V48F and D166Y mutants (with potency D166Y > V48F) led to reduced readily releasable pool (RRP) size, due to increased spontaneous (miniature Excitatory Postsynaptic Current, mEPSC) release and decreased priming rates. These mutations lowered the energy barrier for fusion and increased the release probability, which are gain-of-function features not found in Syt1 knockout (KO) neurons; normalized mEPSC release rates were higher (potency D166Y > V48F) than in the Syt1 KO. These mutations (potency D166Y > V48F) increased spontaneous association to partner SNAREs, resulting in unregulated membrane fusion. In contrast, the I67N mutant decreased mEPSC frequency and evoked EPSC amplitudes due to an increase in the height of the energy barrier for fusion, whereas the RRP size was unaffected. This could be partly compensated by positive charges lowering the energy barrier. Overall, pathogenic mutations in SNAP25 cause complex changes in the energy landscape for priming and fusion.
Collapse
Affiliation(s)
- Anna Kádková
- Department of Neuroscience, University of CopenhagenCopenhagenDenmark
| | | | - Maiken Østergaard
- Department of Neuroscience, University of CopenhagenCopenhagenDenmark
| | - Andrea Malsam
- Heidelberg University Biochemistry CenterHeidelbergDenmark
| | - Jörg Malsam
- Heidelberg University Biochemistry CenterHeidelbergDenmark
| | - Fabio Lolicato
- Heidelberg University Biochemistry CenterHeidelbergDenmark
- Department of Physics, University of HelsinkiHelsinkiFinland
| | - Walter Nickel
- Heidelberg University Biochemistry CenterHeidelbergDenmark
| | | | | |
Collapse
|
6
|
Courtney KC, Mandal T, Mehta N, Wu L, Li Y, Das D, Cui Q, Chapman ER. Synaptotagmin-7 outperforms synaptotagmin-1 to promote the formation of large, stable fusion pores via robust membrane penetration. Nat Commun 2023; 14:7761. [PMID: 38012142 PMCID: PMC10681989 DOI: 10.1038/s41467-023-42497-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 10/11/2023] [Indexed: 11/29/2023] Open
Abstract
Synaptotagmin-1 and synaptotagmin-7 are two prominent calcium sensors that regulate exocytosis in neuronal and neuroendocrine cells. Upon binding calcium, both proteins partially penetrate lipid bilayers that bear anionic phospholipids, but the specific underlying mechanisms that enable them to trigger exocytosis remain controversial. Here, we examine the biophysical properties of these two synaptotagmin isoforms and compare their interactions with phospholipid membranes. We discover that synaptotagmin-1-membrane interactions are greatly influenced by membrane order; tight packing of phosphatidylserine inhibits binding due to impaired membrane penetration. In contrast, synaptotagmin-7 exhibits robust membrane binding and penetration activity regardless of phospholipid acyl chain structure. Thus, synaptotagmin-7 is a super-penetrator. We exploit these observations to specifically isolate and examine the role of membrane penetration in synaptotagmin function. Using nanodisc-black lipid membrane electrophysiology, we demonstrate that membrane penetration is a critical component that underlies how synaptotagmin proteins regulate reconstituted, exocytic fusion pores in response to calcium.
Collapse
Affiliation(s)
- Kevin C Courtney
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Taraknath Mandal
- Department of Chemistry, Boston University, Boston, MA, 02215, USA
- Department of Physics, Indian Institute of Technology - Kanpur, Kanpur, 208016, India
| | - Nikunj Mehta
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Lanxi Wu
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Yueqi Li
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
- Center for Bioanalytical Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Debasis Das
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Navy Nagar, Colaba, Mumbai, 400005, India
| | - Qiang Cui
- Department of Chemistry, Boston University, Boston, MA, 02215, USA
| | - Edwin R Chapman
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA.
| |
Collapse
|
7
|
Norman CA, Krishnakumar SS, Timofeeva Y, Volynski KE. The release of inhibition model reproduces kinetics and plasticity of neurotransmitter release in central synapses. Commun Biol 2023; 6:1091. [PMID: 37891212 PMCID: PMC10611806 DOI: 10.1038/s42003-023-05445-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Calcium-evoked release of neurotransmitters from synaptic vesicles (SVs) is catalysed by SNARE proteins. The predominant view is that, at rest, complete assembly of SNARE complexes is inhibited ('clamped') by synaptotagmin and complexin molecules. Calcium binding by synaptotagmins releases this fusion clamp and triggers fast SV exocytosis. However, this model has not been quantitatively tested over physiological timescales. Here we describe an experimentally constrained computational modelling framework to quantitatively assess how the molecular architecture of the fusion clamp affects SV exocytosis. Our results argue that the 'release-of-inhibition' model can indeed account for fast calcium-activated SV fusion, and that dual binding of synaptotagmin-1 and synaptotagmin-7 to the same SNARE complex enables synergistic regulation of the kinetics and plasticity of neurotransmitter release. The developed framework provides a powerful and adaptable tool to link the molecular biochemistry of presynaptic proteins to physiological data and efficiently test the plausibility of calcium-activated neurotransmitter release models.
Collapse
Affiliation(s)
- Christopher A Norman
- University College London Institute of Neurology, University College London, London, WC1N 3BG, UK
- Department of Computer Science, University of Warwick, Coventry, CV4 7AL, UK
- Mathematics for Real-World Systems Centre for Doctoral Training, University of Warwick, Coventry, CV4 7AL, UK
| | - Shyam S Krishnakumar
- University College London Institute of Neurology, University College London, London, WC1N 3BG, UK.
- Department of Neurology, Yale Nanobiology Institute, Yale University School of Medicine, New Haven, CT, 06510, USA.
| | - Yulia Timofeeva
- University College London Institute of Neurology, University College London, London, WC1N 3BG, UK.
- Department of Computer Science, University of Warwick, Coventry, CV4 7AL, UK.
| | - Kirill E Volynski
- University College London Institute of Neurology, University College London, London, WC1N 3BG, UK.
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
8
|
Warpechowski M, Warpechowski J, Kulczyńska-Przybik A, Mroczko B. Biomarkers of Activity-Dependent Plasticity and Persistent Enhancement of Synaptic Transmission in Alzheimer Disease: A Review of the Current Status. Med Sci Monit 2023; 29:e938826. [PMID: 36600577 PMCID: PMC9832729 DOI: 10.12659/msm.938826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Alzheimer disease (AD) is a chronic and heterogeneous neurodegenerative disorder characterized by complex pathological processes involving neuroinflammation, neurodegeneration, and synaptic dysfunction. Understanding the exact neurobiological mechanisms underlying AD pathology may help to provide a biomarker for early diagnosis or at least for assessment of vulnerability to dementia development. Neural plasticity is defined as a capability of the brain to respond to alterations including aging, injury, or learning, with a crucial role of synaptic elements. Long-term potentiation (LTP) and long-term depression (LTD) are important in regulating synaptic connections between neural cells in functional plasticity. Synaptic loss and impairment of the brain's plasticity in AD leads to cognitive impairment, and one of important roles of synaptic biomarkers is monitoring synaptic dysfunction, response to treatment, and predicting future development of AD. Synaptic biomarkers are undoubtedly very promising in developing novel approach to AD treatment and control, especially in the era of aging of societies, which is one of the most common risk factor of AD. Implementing a widespread measurement of synaptic biomarkers of AD will probably be crucial in early diagnosis of AD, early therapeutic intervention, monitoring progression of the disease, or response to treatment. One of the most important challenges is finding a biomarker whose blood concentration correlates with its level in the central nervous system (CNS). This review aims to present the current status of biomarkers of activity-dependent plasticity and persistent enhancement of synaptic transmission in Alzheimer disease.
Collapse
Affiliation(s)
- Marcin Warpechowski
- Department of Statistics and Medical Informatics, Medical University of Białystok, Białystok, Poland
| | | | | | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, Białystok, Poland,Department of Biochemical Diagnostics, University Hospital of Białystok, Białystok, Poland
| |
Collapse
|
9
|
Jaczynska K, Esquivies L, Pfuetzner RA, Alten B, Brewer KD, Zhou Q, Kavalali ET, Brunger AT, Rizo J. Analysis of tripartite Synaptotagmin-1-SNARE-complexin-1 complexes in solution. FEBS Open Bio 2023; 13:26-50. [PMID: 36305864 PMCID: PMC9811660 DOI: 10.1002/2211-5463.13503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/19/2022] [Accepted: 10/27/2022] [Indexed: 01/07/2023] Open
Abstract
Characterizing interactions of Synaptotagmin-1 with the SNARE complex is crucial to understand the mechanism of neurotransmitter release. X-ray crystallography revealed how the Synaptotagmin-1 C2 B domain binds to the SNARE complex through a so-called primary interface and to a complexin-1-SNARE complex through a so-called tripartite interface. Mutagenesis and electrophysiology supported the functional relevance of both interfaces, and extensive additional data validated the primary interface. However, ITC evidence suggesting that binding via the tripartite interface occurs in solution was called into question by subsequent NMR data. Here, we describe joint efforts to address this apparent contradiction. Using the same ITC approach with the same C2 B domain mutant used previously (C2 BKA-Q ) but including ion exchange chromatography to purify it, which is crucial to remove polyacidic contaminants, we were unable to observe the substantial endothermic ITC signal that was previously attributed to binding of this mutant to the complexin-1-SNARE complex through the tripartite interface. We were also unable to detect substantial populations of the tripartite interface in NMR analyses of the ITC samples or in measurements of paramagnetic relaxation effects, despite the high sensitivity of this method to detect weak protein complexes. However, these experiments do not rule out the possibility of very low affinity (KD > 1 mm) binding through this interface. These results emphasize the need to develop methods to characterize the structure of synaptotagmin-1-SNARE complexes between two membranes and to perform further structure-function analyses to establish the physiological relevance of the tripartite interface.
Collapse
Affiliation(s)
- Klaudia Jaczynska
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Department of BiochemistryUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Department of PharmacologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Luis Esquivies
- Department of Molecular and Cellular PhysiologyStanford UniversityCAUSA
- Department of Neurology and Neurological SciencesStanford UniversityCAUSA
- Department of Structural BiologyStanford UniversityCAUSA
- Department of Photon ScienceStanford UniversityCAUSA
- Howard Hughes Medical InstituteStanford UniversityCAUSA
| | - Richard A. Pfuetzner
- Department of Molecular and Cellular PhysiologyStanford UniversityCAUSA
- Department of Neurology and Neurological SciencesStanford UniversityCAUSA
- Department of Structural BiologyStanford UniversityCAUSA
- Department of Photon ScienceStanford UniversityCAUSA
- Howard Hughes Medical InstituteStanford UniversityCAUSA
| | - Baris Alten
- Department of PharmacologyVanderbilt UniversityNashvilleTNUSA
- Vanderbilt Brain InstituteVanderbilt UniversityNashvilleTNUSA
- Present address:
Department of NeurologyMassachusetts General HospitalBostonMAUSA
- Present address:
Department of NeurologyBrigham and Women's HospitalBostonMAUSA
- Present address:
Harvard Medical SchoolBostonMAUSA
| | - Kyle D. Brewer
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Department of BiochemistryUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Department of PharmacologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Present address:
ETTA BiotechnologyPalo AltoCAUSA
| | - Qiangjun Zhou
- Vanderbilt Brain InstituteVanderbilt UniversityNashvilleTNUSA
- Department of Cell and Developmental BiologyVanderbilt UniversityNashvilleTNUSA
| | - Ege T. Kavalali
- Department of PharmacologyVanderbilt UniversityNashvilleTNUSA
- Vanderbilt Brain InstituteVanderbilt UniversityNashvilleTNUSA
| | - Axel T. Brunger
- Department of Molecular and Cellular PhysiologyStanford UniversityCAUSA
- Department of Neurology and Neurological SciencesStanford UniversityCAUSA
- Department of Structural BiologyStanford UniversityCAUSA
- Department of Photon ScienceStanford UniversityCAUSA
- Howard Hughes Medical InstituteStanford UniversityCAUSA
| | - Josep Rizo
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Department of BiochemistryUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Department of PharmacologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
| |
Collapse
|
10
|
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: 2.0] [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
|
11
|
Zhou Q. Calcium Sensors of Neurotransmitter Release. ADVANCES IN NEUROBIOLOGY 2023; 33:119-138. [PMID: 37615865 DOI: 10.1007/978-3-031-34229-5_5] [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
Calcium (Ca2+) plays a critical role in triggering all three primary modes of neurotransmitter release (synchronous, asynchronous, and spontaneous). Synaptotagmin1, a protein with two C2 domains, is the first isoform of the synaptotagmin family that was identified and demonstrated as the primary Ca2+ sensor for synchronous neurotransmitter release. Other isoforms of the synaptotagmin family as well as other C2 proteins such as the double C2 domain protein family were found to act as Ca2+ sensors for different modes of neurotransmitter release. Major recent advances and previous data suggest a new model, release-of-inhibition, for the initiation of Ca2+-triggered synchronous neurotransmitter release. Synaptotagmin1 binds Ca2+ via its two C2 domains and relieves a primed pre-fusion machinery. Before Ca2+ triggering, synaptotagmin1 interacts Ca2+ independently with partially zippered SNARE complexes, the plasma membrane, phospholipids, and other components to form a primed pre-fusion state that is ready for fast release. However, membrane fusion is inhibited until the arrival of Ca2+ reorients the Ca2+-binding loops of the C2 domain to perturb the lipid bilayers, help bridge the membranes, and/or induce membrane curvatures, which serves as a power stroke to activate fusion. This chapter reviews the evidence supporting these models and discusses the molecular interactions that may underlie these abilities.
Collapse
Affiliation(s)
- Qiangjun Zhou
- Department of Cell and Developmental Biology, Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
12
|
Rizo J, David G, Fealey ME, Jaczynska K. On the difficulties of characterizing weak protein interactions that are critical for neurotransmitter release. FEBS Open Bio 2022; 12:1912-1938. [PMID: 35986639 PMCID: PMC9623538 DOI: 10.1002/2211-5463.13473] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/09/2022] [Accepted: 08/18/2022] [Indexed: 01/25/2023] Open
Abstract
The mechanism of neurotransmitter release has been extensively characterized, showing that vesicle fusion is mediated by the SNARE complex formed by syntaxin-1, SNAP-25 and synaptobrevin. This complex is disassembled by N-ethylmaleimide sensitive factor (NSF) and SNAPs to recycle the SNAREs, whereas Munc18-1 and Munc13s organize SNARE complex assembly in an NSF-SNAP-resistant manner. Synaptotagmin-1 acts as the Ca2+ sensor that triggers exocytosis in a tight interplay with the SNAREs and complexins. Here, we review technical aspects associated with investigation of protein interactions underlying these steps, which is hindered because the release machinery is assembled between two membranes and is highly dynamic. Moreover, weak interactions, which are difficult to characterize, play key roles in neurotransmitter release, for instance by lowering energy barriers that need to be overcome in this highly regulated process. We illustrate the crucial role that structural biology has played in uncovering mechanisms underlying neurotransmitter release, but also discuss the importance of considering the limitations of the techniques used, including lessons learned from research in our lab and others. In particular, we emphasize: (a) the promiscuity of some protein sequences, including membrane-binding regions that can mediate irrelevant interactions with proteins in the absence of their native targets; (b) the need to ensure that weak interactions observed in crystal structures are biologically relevant; and (c) the limitations of isothermal titration calorimetry to analyze weak interactions. Finally, we stress that even studies that required re-interpretation often helped to move the field forward by improving our understanding of the system and providing testable hypotheses.
Collapse
Affiliation(s)
- Josep Rizo
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTXUSA,Department of BiochemistryUniversity of Texas Southwestern Medical CenterDallasTXUSA,Department of PharmacologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Guillaume David
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTXUSA,Department of BiochemistryUniversity of Texas Southwestern Medical CenterDallasTXUSA,Department of PharmacologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Michael E. Fealey
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTXUSA,Department of BiochemistryUniversity of Texas Southwestern Medical CenterDallasTXUSA,Department of PharmacologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Klaudia Jaczynska
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTXUSA,Department of BiochemistryUniversity of Texas Southwestern Medical CenterDallasTXUSA,Department of PharmacologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
| |
Collapse
|
13
|
Rizo J, Sari L, Qi Y, Im W, Lin MM. All-atom molecular dynamics simulations of Synaptotagmin-SNARE-complexin complexes bridging a vesicle and a flat lipid bilayer. eLife 2022; 11:76356. [PMID: 35708237 PMCID: PMC9239685 DOI: 10.7554/elife.76356] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 06/15/2022] [Indexed: 12/14/2022] Open
Abstract
Synaptic vesicles are primed into a state that is ready for fast neurotransmitter release upon Ca2+-binding to Synaptotagmin-1. This state likely includes trans-SNARE complexes between the vesicle and plasma membranes that are bound to Synaptotagmin-1 and complexins. However, the nature of this state and the steps leading to membrane fusion are unclear, in part because of the difficulty of studying this dynamic process experimentally. To shed light into these questions, we performed all-atom molecular dynamics simulations of systems containing trans-SNARE complexes between two flat bilayers or a vesicle and a flat bilayer with or without fragments of Synaptotagmin-1 and/or complexin-1. Our results need to be interpreted with caution because of the limited simulation times and the absence of key components, but suggest mechanistic features that may control release and help visualize potential states of the primed Synaptotagmin-1-SNARE-complexin-1 complex. The simulations suggest that SNAREs alone induce formation of extended membrane-membrane contact interfaces that may fuse slowly, and that the primed state contains macromolecular assemblies of trans-SNARE complexes bound to the Synaptotagmin-1 C2B domain and complexin-1 in a spring-loaded configuration that prevents premature membrane merger and formation of extended interfaces, but keeps the system ready for fast fusion upon Ca2+ influx.
Collapse
Affiliation(s)
- Josep Rizo
- Department of Biophysics, The 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
| | - Levent Sari
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, United States.,Green Center for Systems Biology, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Yife Qi
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Wonpil Im
- Department of Biological Sciences, Lehigh University, Bethlehem, United States.,Department of Chemistry, Lehigh University, Bethlehem, United States.,Department of Bioengineering, Lehigh University, Bethlehem, United States.,Department of Computer Science and Engineering, Lehigh University, Bethlehem, United States
| | - Milo M Lin
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, United States.,Green Center for Systems Biology, The University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
14
|
Abstract
Major recent advances and previous data have led to a plausible model of how key proteins mediate neurotransmitter release. In this model, the soluble N-ethylmaleimide-sensitive factor (NSF) attachment protein (SNAP) receptor (SNARE) proteins syntaxin-1, SNAP-25, and synaptobrevin form tight complexes that bring the membranes together and are crucial for membrane fusion. NSF and SNAPs disassemble SNARE complexes and ensure that fusion occurs through an exquisitely regulated pathway that starts with Munc18-1 bound to a closed conformation of syntaxin-1. Munc18-1 also binds to synaptobrevin, forming a template to assemble the SNARE complex when Munc13-1 opens syntaxin-1 while bridging the vesicle and plasma membranes. Synaptotagmin-1 and complexin bind to partially assembled SNARE complexes, likely stabilizing them and preventing fusion until Ca2+ binding to synaptotagmin-1 causes dissociation from the SNARE complex and induces interactions with phospholipids that help trigger release. Although fundamental questions remain about the mechanism of membrane fusion, these advances provide a framework to investigate the mechanisms underlying presynaptic plasticity.
Collapse
Affiliation(s)
- Josep Rizo
- Departments of Biophysics, Biochemistry, and Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA;
| |
Collapse
|
15
|
Camacho M, Quade B, Trimbuch T, Xu J, Sari L, Rizo J, Rosenmund C. Control of neurotransmitter release by two distinct membrane-binding faces of the Munc13-1 C 1C 2B region. eLife 2021; 10:e72030. [PMID: 34779770 PMCID: PMC8648301 DOI: 10.7554/elife.72030] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/14/2021] [Indexed: 11/23/2022] Open
Abstract
Munc13-1 plays a central role in neurotransmitter release through its conserved C-terminal region, which includes a diacyglycerol (DAG)-binding C1 domain, a Ca2+/PIP2-binding C2B domain, a MUN domain and a C2C domain. Munc13-1 was proposed to bridge synaptic vesicles to the plasma membrane through distinct interactions of the C1C2B region with the plasma membrane: (i) one involving a polybasic face that is expected to yield a perpendicular orientation of Munc13-1 and hinder release; and (ii) another involving the DAG-Ca2+-PIP2-binding face that is predicted to result in a slanted orientation and facilitate release. Here, we have tested this model and investigated the role of the C1C2B region in neurotransmitter release. We find that K603E or R769E point mutations in the polybasic face severely impair Ca2+-independent liposome bridging and fusion in in vitro reconstitution assays, and synaptic vesicle priming in primary murine hippocampal cultures. A K720E mutation in the polybasic face and a K706E mutation in the C2B domain Ca2+-binding loops have milder effects in reconstitution assays and do not affect vesicle priming, but enhance or impair Ca2+-evoked release, respectively. The phenotypes caused by combining these mutations are dominated by the K603E and R769E mutations. Our results show that the C1-C2B region of Munc13-1 plays a central role in vesicle priming and support the notion that two distinct faces of this region control neurotransmitter release and short-term presynaptic plasticity.
Collapse
Affiliation(s)
- Marcial Camacho
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of NeurophysiologyBerlinGermany
- NeuroCure Cluster of ExcellenceBerlinGermany
| | - Bradley Quade
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Biochemistry, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Pharmacology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Thorsten Trimbuch
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of NeurophysiologyBerlinGermany
- NeuroCure Cluster of ExcellenceBerlinGermany
| | - Junjie Xu
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Biochemistry, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Pharmacology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Levent Sari
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States
- Cecil H. and Ida Green Comprehensive Center for Molecular, Computational and Systems Biology, University of Texas Southwestern Medical CenterDallasUnited States
- Center for Alzheimer’s and Neurodegenerative Diseases, University of Texas Southwestern Medical CenterDallasUnited States
| | - Josep Rizo
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Biochemistry, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Pharmacology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Christian Rosenmund
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of NeurophysiologyBerlinGermany
- NeuroCure Cluster of ExcellenceBerlinGermany
| |
Collapse
|
16
|
Wu Z, Dharan N, McDargh ZA, Thiyagarajan S, O'Shaughnessy B, Karatekin E. The neuronal calcium sensor Synaptotagmin-1 and SNARE proteins cooperate to dilate fusion pores. eLife 2021; 10:68215. [PMID: 34190041 PMCID: PMC8294851 DOI: 10.7554/elife.68215] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/29/2021] [Indexed: 02/07/2023] Open
Abstract
All membrane fusion reactions proceed through an initial fusion pore, including calcium-triggered release of neurotransmitters and hormones. Expansion of this small pore to release cargo is energetically costly and regulated by cells, but the mechanisms are poorly understood. Here, we show that the neuronal/exocytic calcium sensor Synaptotagmin-1 (Syt1) promotes expansion of fusion pores induced by SNARE proteins. Pore dilation relied on calcium-induced insertion of the tandem C2 domain hydrophobic loops of Syt1 into the membrane, previously shown to reorient the C2 domain. Mathematical modelling suggests that C2B reorientation rotates a bound SNARE complex so that it exerts force on the membranes in a mechanical lever action that increases the height of the fusion pore, provoking pore dilation to offset the bending energy penalty. We conclude that Syt1 exerts novel non-local calcium-dependent mechanical forces on fusion pores that dilate pores and assist neurotransmitter and hormone release.
Collapse
Affiliation(s)
- Zhenyong Wu
- Department of Cellular and Molecular Physiology, Yale University, New Haven, United States.,Nanobiology Institute, Yale University, West Haven, United States
| | - Nadiv Dharan
- Department of Chemical Engineering, Columbia University, New York, United States
| | - Zachary A McDargh
- Department of Chemical Engineering, Columbia University, New York, United States
| | - Sathish Thiyagarajan
- Department of Chemical Engineering, Columbia University, New York, United States
| | - Ben O'Shaughnessy
- Department of Chemical Engineering, Columbia University, New York, United States
| | - Erdem Karatekin
- Department of Cellular and Molecular Physiology, Yale University, New Haven, United States.,Nanobiology Institute, Yale University, West Haven, United States.,Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, United States.,Saints-Pères Paris Institute for the Neurosciences (SPPIN), Université de Paris, Centre National de la Recherche Scientifique (CNRS) UMR 8003, Paris, France
| |
Collapse
|
17
|
Synaptotagmin-1-, Munc18-1-, and Munc13-1-dependent liposome fusion with a few neuronal SNAREs. Proc Natl Acad Sci U S A 2021; 118:2019314118. [PMID: 33468652 DOI: 10.1073/pnas.2019314118] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Neurotransmitter release is governed by eight central proteins among other factors: the neuronal SNAREs syntaxin-1, synaptobrevin, and SNAP-25, which form a tight SNARE complex that brings the synaptic vesicle and plasma membranes together; NSF and SNAPs, which disassemble SNARE complexes; Munc18-1 and Munc13-1, which organize SNARE complex assembly; and the Ca2+ sensor synaptotagmin-1. Reconstitution experiments revealed that Munc18-1, Munc13-1, NSF, and α-SNAP can mediate Ca2+-dependent liposome fusion between synaptobrevin liposomes and syntaxin-1-SNAP-25 liposomes, but high fusion efficiency due to uncontrolled SNARE complex assembly did not allow investigation of the role of synaptotagmin-1 on fusion. Here, we show that decreasing the synaptobrevin-to-lipid ratio in the corresponding liposomes to very low levels leads to inefficient fusion and that synaptotagmin-1 strongly stimulates fusion under these conditions. Such stimulation depends on Ca2+ binding to the two C2 domains of synaptotagmin-1. We also show that anchoring SNAP-25 on the syntaxin-1 liposomes dramatically enhances fusion. Moreover, we uncover a synergy between synaptotagmin-1 and membrane anchoring of SNAP-25, which allows efficient Ca2+-dependent fusion between liposomes bearing very low synaptobrevin densities and liposomes containing very low syntaxin-1 densities. Thus, liposome fusion in our assays is achieved with a few SNARE complexes in a manner that requires Munc18-1 and Munc13-1 and that depends on Ca2+ binding to synaptotagmin-1, all of which are fundamental features of neurotransmitter release in neurons.
Collapse
|
18
|
Uchino K, Kawano H, Tanaka Y, Adaniya Y, Asahara A, Deshimaru M, Kubota K, Watanabe T, Katsurabayashi S, Iwasaki K, Hirose S. Inhibitory synaptic transmission is impaired at higher extracellular Ca 2+ concentrations in Scn1a +/- mouse model of Dravet syndrome. Sci Rep 2021; 11:10634. [PMID: 34017040 PMCID: PMC8137694 DOI: 10.1038/s41598-021-90224-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022] Open
Abstract
Dravet syndrome (DS) is an intractable form of childhood epilepsy that occurs in infancy. More than 80% of all patients have a heterozygous abnormality in the SCN1A gene, which encodes a subunit of Na+ channels in the brain. However, the detailed pathogenesis of DS remains unclear. This study investigated the synaptic pathogenesis of this disease in terms of excitatory/inhibitory balance using a mouse model of DS. We show that excitatory postsynaptic currents were similar between Scn1a knock-in neurons (Scn1a+/- neurons) and wild-type neurons, but inhibitory postsynaptic currents were significantly lower in Scn1a+/- neurons. Moreover, both the vesicular release probability and the number of inhibitory synapses were significantly lower in Scn1a+/- neurons compared with wild-type neurons. There was no proportional increase in inhibitory postsynaptic current amplitude in response to increased extracellular Ca2+ concentrations. Our study revealed that the number of inhibitory synapses is significantly reduced in Scn1a+/- neurons, while the sensitivity of inhibitory synapses to extracellular Ca2+ concentrations is markedly increased. These data suggest that Ca2+ tethering in inhibitory nerve terminals may be disturbed following the synaptic burst, likely leading to epileptic symptoms.
Collapse
Affiliation(s)
- Kouya Uchino
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Hiroyuki Kawano
- Research Institute for the Molecular Pathogeneses of Epilepsy, Fukuoka University, Fukuoka, Japan
| | - Yasuyoshi Tanaka
- Research Institute for the Molecular Pathogeneses of Epilepsy, Fukuoka University, Fukuoka, Japan
| | - Yuna Adaniya
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Ai Asahara
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Masanobu Deshimaru
- Research Institute for the Molecular Pathogeneses of Epilepsy, Fukuoka University, Fukuoka, Japan
| | - Kaori Kubota
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Takuya Watanabe
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Shutaro Katsurabayashi
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan.
- Research Institute for the Molecular Pathogeneses of Epilepsy, Fukuoka University, Fukuoka, Japan.
| | - Katsunori Iwasaki
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Shinichi Hirose
- Research Institute for the Molecular Pathogeneses of Epilepsy, Fukuoka University, Fukuoka, Japan
- Department of Pediatrics, School of Medicine, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
19
|
Thoreson WB. Transmission at rod and cone ribbon synapses in the retina. Pflugers Arch 2021; 473:1469-1491. [PMID: 33779813 DOI: 10.1007/s00424-021-02548-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/29/2022]
Abstract
Light-evoked voltage responses of rod and cone photoreceptor cells in the vertebrate retina must be converted to a train of synaptic vesicle release events for transmission to downstream neurons. This review discusses the processes, proteins, and structures that shape this critical early step in vision, focusing on studies from salamander retina with comparisons to other experimental animals. Many mechanisms are conserved across species. In cones, glutamate release is confined to ribbon release sites although rods are also capable of release at non-ribbon sites. The role of non-ribbon release in rods remains unclear. Release from synaptic ribbons in rods and cones involves at least three vesicle pools: a readily releasable pool (RRP) matching the number of membrane-associated vesicles along the ribbon base, a ribbon reserve pool matching the number of additional vesicles on the ribbon, and an enormous cytoplasmic reserve. Vesicle release increases in parallel with Ca2+ channel activity. While the opening of only a few Ca2+ channels beneath each ribbon can trigger fusion of a single vesicle, sustained release rates in darkness are governed by the rate at which the RRP can be replenished. The number of vacant release sites, their functional status, and the rate of vesicle delivery in turn govern replenishment. Along with an overview of the mechanisms of exocytosis and endocytosis, we consider specific properties of ribbon-associated proteins and pose a number of remaining questions about this first synapse in the visual system.
Collapse
Affiliation(s)
- Wallace B Thoreson
- Truhlsen Eye Institute, Departments of Ophthalmology & Visual Sciences and Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
20
|
Lee JH, Kim JY, Noh S, Lee H, Lee SY, Mun JY, Park H, Chung WS. Astrocytes phagocytose adult hippocampal synapses for circuit homeostasis. Nature 2020; 590:612-617. [PMID: 33361813 DOI: 10.1038/s41586-020-03060-3] [Citation(s) in RCA: 177] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023]
Abstract
In the adult hippocampus, synapses are constantly formed and eliminated1,2. However, the exact function of synapse elimination in the adult brain, and how it is regulated, are largely unknown. Here we show that astrocytic phagocytosis3 is important for maintaining proper hippocampal synaptic connectivity and plasticity. By using fluorescent phagocytosis reporters, we find that excitatory and inhibitory synapses are eliminated by glial phagocytosis in the CA1 region of the adult mouse hippocampus. Unexpectedly, we found that astrocytes have a major role in the neuronal activity-dependent elimination of excitatory synapses. Furthermore, mice in which astrocytes lack the phagocytic receptor MEGF10 show a reduction in the elimination of excitatory synapses; as a result, excessive but functionally impaired synapses accumulate. Finally, Megf10-knockout mice show defective long-term synaptic plasticity and impaired formation of hippocampal memories. Together, our data provide strong evidence that astrocytes eliminate unnecessary excitatory synaptic connections in the adult hippocampus through MEGF10, and that this astrocytic function is crucial for maintaining circuit connectivity and thereby supporting cognitive function.
Collapse
Affiliation(s)
- Joon-Hyuk Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ji-Young Kim
- Research Group for Neurovascular Unit, Korea Brain Research Institute (KBRI), Daegu, Republic of Korea.,Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Seulgi Noh
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.,Research Group for Neural Circuit, Korea Brain Research Institute (KBRI), Daegu, Republic of Korea
| | - Hyoeun Lee
- Research Group for Neurovascular Unit, Korea Brain Research Institute (KBRI), Daegu, Republic of Korea
| | - Se Young Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ji Young Mun
- Research Group for Neural Circuit, Korea Brain Research Institute (KBRI), Daegu, Republic of Korea
| | - Hyungju Park
- Research Group for Neurovascular Unit, Korea Brain Research Institute (KBRI), Daegu, Republic of Korea. .,Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.
| | - Won-Suk Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
| |
Collapse
|
21
|
Caparotta M, Tomes CN, Mayorga LS, Masone D. The Synaptotagmin-1 C2B Domain Is a Key Regulator in the Stabilization of the Fusion Pore. J Chem Theory Comput 2020; 16:7840-7851. [DOI: 10.1021/acs.jctc.0c00734] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Marcelo Caparotta
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo (UNCuyo), Mendoza 5500, Argentina
| | - Claudia N. Tomes
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo (UNCuyo), Mendoza 5500, Argentina
- Instituto de Histología y Embriología de Mendoza (IHEM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo (UNCuyo), Mendoza 5500, Argentina
| | - Luis S. Mayorga
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo (UNCuyo), Mendoza 5500, Argentina
- Instituto de Histología y Embriología de Mendoza (IHEM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo (UNCuyo), Mendoza 5500, Argentina
| | - Diego Masone
- Instituto de Histología y Embriología de Mendoza (IHEM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo (UNCuyo), Mendoza 5500, Argentina
- Facultad de Ingeniería, Universidad Nacional de Cuyo (UNCuyo), Mendoza 5500, Argentina
| |
Collapse
|
22
|
Huson V, Meijer M, Dekker R, Ter Veer M, Ruiter M, van Weering JR, Verhage M, Cornelisse LN. Post-tetanic potentiation lowers the energy barrier for synaptic vesicle fusion independently of Synaptotagmin-1. eLife 2020; 9:55713. [PMID: 32831174 PMCID: PMC7500951 DOI: 10.7554/elife.55713] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 08/23/2020] [Indexed: 11/13/2022] Open
Abstract
Previously, we showed that modulation of the energy barrier for synaptic vesicle fusion boosts release rates supralinearly (Schotten, 2015). Here we show that mouse hippocampal synapses employ this principle to trigger Ca2+-dependent vesicle release and post-tetanic potentiation (PTP). We assess energy barrier changes by fitting release kinetics in response to hypertonic sucrose. Mimicking activation of the C2A domain of the Ca2+-sensor Synaptotagmin-1 (Syt1), by adding a positive charge (Syt1D232N) or increasing its hydrophobicity (Syt14W), lowers the energy barrier. Removing Syt1 or impairing its release inhibitory function (Syt19Pro) increases spontaneous release without affecting the fusion barrier. Both phorbol esters and tetanic stimulation potentiate synaptic strength, and lower the energy barrier equally well in the presence and absence of Syt1. We propose a model where tetanic stimulation activates Syt1-independent mechanisms that lower the energy barrier and act additively with Syt1-dependent mechanisms to produce PTP by exerting multiplicative effects on release rates.
Collapse
Affiliation(s)
- Vincent Huson
- Department of Functional Genomics, Clinical Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center- Location VUmc, Amsterdam, Netherlands
| | - Marieke Meijer
- Department of Functional Genomics, Clinical Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center- Location VUmc, Amsterdam, Netherlands
| | - Rien Dekker
- Department of Functional Genomics, Clinical Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center- Location VUmc, Amsterdam, Netherlands
| | - Mirelle Ter Veer
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, Netherlands
| | - Marvin Ruiter
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, Netherlands
| | - Jan Rt van Weering
- Department of Functional Genomics, Clinical Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center- Location VUmc, Amsterdam, Netherlands
| | - Matthijs Verhage
- Department of Functional Genomics, Clinical Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center- Location VUmc, Amsterdam, Netherlands.,Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, Netherlands
| | - Lennart Niels Cornelisse
- Department of Functional Genomics, Clinical Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center- Location VUmc, Amsterdam, Netherlands
| |
Collapse
|
23
|
Voleti R, Jaczynska K, Rizo J. Ca 2+-dependent release of synaptotagmin-1 from the SNARE complex on phosphatidylinositol 4,5-bisphosphate-containing membranes. eLife 2020; 9:57154. [PMID: 32808925 PMCID: PMC7498268 DOI: 10.7554/elife.57154] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 08/12/2020] [Indexed: 11/13/2022] Open
Abstract
The Ca2+ sensor synaptotagmin-1 and the SNARE complex cooperate to trigger neurotransmitter release. Structural studies elucidated three distinct synaptotagmin-1-SNARE complex binding modes involving 'polybasic', 'primary' and 'tripartite' interfaces of synaptotagmin-1. We investigated these interactions using NMR and fluorescence spectroscopy. Synaptotagmin-1 binds to the SNARE complex through the polybasic and primary interfaces in solution. Ca2+-free synaptotagmin-1 binds to SNARE complexes anchored on PIP2-containing nanodiscs. R398Q/R399Q and E295A/Y338W mutations at the primary interface, which strongly impair neurotransmitter release, disrupt and enhance synaptotagmin-1-SNARE complex binding, respectively. Ca2+ induces tight binding of synaptotagmin-1 to PIP2-containing nanodiscs, disrupting synaptotagmin-1-SNARE interactions. Specific effects of mutations in the polybasic region on Ca2+-dependent synaptotagmin-1-PIP2-membrane interactions correlate with their effects on release. Our data suggest that synaptotagmin-1 binds to the SNARE complex through the primary interface and that Ca2+ releases this interaction, inducing PIP2/membrane binding and allowing cooperation between synaptotagmin-1 and the SNAREs in membrane fusion to trigger release.
Collapse
Affiliation(s)
- Rashmi Voleti
- 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
| | - Klaudia Jaczynska
- 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
| | - 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
|
24
|
Bonnycastle K, Davenport EC, Cousin MA. Presynaptic dysfunction in neurodevelopmental disorders: Insights from the synaptic vesicle life cycle. J Neurochem 2020; 157:179-207. [PMID: 32378740 DOI: 10.1111/jnc.15035] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/14/2020] [Accepted: 04/22/2020] [Indexed: 12/11/2022]
Abstract
The activity-dependent fusion, retrieval and recycling of synaptic vesicles is essential for the maintenance of neurotransmission. Until relatively recently it was believed that most mutations in genes that were essential for this process would be incompatible with life, because of this fundamental role. However, an ever-expanding number of mutations in this very cohort of genes are being identified in individuals with neurodevelopmental disorders, including autism, intellectual disability and epilepsy. This article will summarize the current state of knowledge linking mutations in presynaptic genes to neurodevelopmental disorders by sequentially covering the various stages of the synaptic vesicle life cycle. It will also discuss how perturbations of specific stages within this recycling process could translate into human disease. Finally, it will also provide perspectives on the potential for future therapy that are targeted to presynaptic function.
Collapse
Affiliation(s)
- Katherine Bonnycastle
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.,Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Elizabeth C Davenport
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.,Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Michael A Cousin
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.,Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
25
|
Ruiter M, Kádková A, Scheutzow A, Malsam J, Söllner TH, Sørensen JB. An Electrostatic Energy Barrier for SNARE-Dependent Spontaneous and Evoked Synaptic Transmission. Cell Rep 2020; 26:2340-2352.e5. [PMID: 30811985 DOI: 10.1016/j.celrep.2019.01.103] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 11/05/2018] [Accepted: 01/28/2019] [Indexed: 12/22/2022] Open
Abstract
Information transfer across CNS synapses depends on the very low basal vesicle fusion rate and the ability to rapidly upregulate that rate upon Ca2+ influx. We show that local electrostatic repulsion participates in creating an energy barrier, which limits spontaneous synaptic transmission. The barrier amplitude is increased by negative charges and decreased by positive charges on the SNARE-complex surface. Strikingly, the effect of charges on the barrier is additive and this extends to evoked transmission, but with a shallower charge dependence. Action potential-driven synaptic release is equivalent to the abrupt addition of ∼35 positive charges to the fusion machine. Within an electrostatic model for triggering, the Ca2+ sensor synaptotagmin-1 contributes ∼18 charges by binding Ca2+, while also modulating the fusion barrier at rest. Thus, the energy barrier for synaptic vesicle fusion has a large electrostatic component, allowing synaptotagmin-1 to act as an electrostatic switch and modulator to trigger vesicle fusion.
Collapse
Affiliation(s)
- Marvin Ruiter
- Department of Neuroscience, Faculty of Health and Medical Sciences, 2200 Copenhagen N, University of Copenhagen, Copenhagen, Denmark
| | - Anna Kádková
- Department of Neuroscience, Faculty of Health and Medical Sciences, 2200 Copenhagen N, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Scheutzow
- Heidelberg University Biochemistry Center, 69120 Heidelberg, Germany
| | - Jörg Malsam
- Heidelberg University Biochemistry Center, 69120 Heidelberg, Germany
| | - Thomas H Söllner
- Heidelberg University Biochemistry Center, 69120 Heidelberg, Germany
| | - Jakob B Sørensen
- Department of Neuroscience, Faculty of Health and Medical Sciences, 2200 Copenhagen N, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
26
|
Bowers MR, Reist NE. Synaptotagmin: Mechanisms of an electrostatic switch. Neurosci Lett 2020; 722:134834. [DOI: 10.1016/j.neulet.2020.134834] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/06/2020] [Accepted: 02/09/2020] [Indexed: 02/09/2023]
|
27
|
Bowers MR, Reist NE. The C2A domain of synaptotagmin is an essential component of the calcium sensor for synaptic transmission. PLoS One 2020; 15:e0228348. [PMID: 32032373 PMCID: PMC7006929 DOI: 10.1371/journal.pone.0228348] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/13/2020] [Indexed: 01/27/2023] Open
Abstract
The synaptic vesicle protein, synaptotagmin, is the principle Ca2+ sensor for synaptic transmission. Ca2+ influx into active nerve terminals is translated into neurotransmitter release by Ca2+ binding to synaptotagmin’s tandem C2 domains, triggering the fast, synchronous fusion of multiple synaptic vesicles. Two hydrophobic residues, shown to mediate Ca2+-dependent membrane insertion of these C2 domains, are required for this process. Previous research suggested that one of its tandem C2 domains (C2B) is critical for fusion, while the other domain (C2A) plays only a facilitatory role. However, the function of the two hydrophobic residues in C2A have not been adequately tested in vivo. Here we show that these two hydrophobic residues are absolutely required for synaptotagmin to trigger vesicle fusion. Using in vivo electrophysiological recording at the Drosophila larval neuromuscular junction, we found that mutation of these two key C2A hydrophobic residues almost completely abolished neurotransmitter release. Significantly, mutation of both hydrophobic residues resulted in more severe deficits than those seen in synaptotagmin null mutants. Thus, we report the most severe phenotype of a C2A mutation to date, demonstrating that the C2A domain is absolutely essential for synaptotagmin’s function as the electrostatic switch. The postulated role of synaptotagmin’s C2A domain in triggering neurotransmitter release has fluctuated wildly over the years. Early biochemical experiments suggested that the C2A domain was essential, while the C2B domain was superfluous. Then, functional experiments measuring neurotransmitter release in vivo following disruptions in Ca2+ binding suggested that C2B was essential, while C2A was superfluous. Subsequently, the use of more refined mutations to disrupt Ca2+ binding indicated that C2A played a facilitatory role. Here we show two hydrophobic residues of the C2A domain are absolutely required for synaptotagmin-triggered neurotransmitter release. Thus, after over twenty years of research, we now demonstrate that the C2A domain of synaptotagmin is an essential component of the Ca2+ sensor for triggering synaptic transmission in vivo.
Collapse
Affiliation(s)
- Matthew R. Bowers
- Department of Biomedical Sciences, Molecular, Cellular, Integrative Neurosciences Program, Colorado State University, Fort Collins, CO, United States of America
| | - Noreen E. Reist
- Department of Biomedical Sciences, Molecular, Cellular, Integrative Neurosciences Program, Colorado State University, Fort Collins, CO, United States of America
- * E-mail:
| |
Collapse
|
28
|
Synaptotagmin 1 oligomers clamp and regulate different modes of neurotransmitter release. Proc Natl Acad Sci U S A 2020; 117:3819-3827. [PMID: 32015138 PMCID: PMC7035618 DOI: 10.1073/pnas.1920403117] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Release of neurotransmitters relies on submillisecond coupling of synaptic vesicle fusion to the triggering signal: AP-evoked presynaptic Ca2+ influx. The key player that controls exocytosis of the synaptic vesicle is the Ca2+ sensor synaptotagmin 1 (Syt1). While the Ca2+ activation of Syt1 has been extensively characterized, how Syt1 reversibly clamps vesicular fusion remains enigmatic. Here, using a targeted mutation combined with fluorescence imaging and electrophysiology, we show that the structural feature of Syt1 to self-oligomerize provides the molecular basis for clamping of spontaneous and asynchronous release but is not required for triggering of synchronous release. Our findings propose a mechanistic model that explains how Syt1 oligomers regulate different modes of transmitter release in neuronal synapses. Synaptotagmin 1 (Syt1) synchronizes neurotransmitter release to action potentials (APs) acting as the fast Ca2+ release sensor and as the inhibitor (clamp) of spontaneous and delayed asynchronous release. While the Syt1 Ca2+ activation mechanism has been well-characterized, how Syt1 clamps transmitter release remains enigmatic. Here we show that C2B domain-dependent oligomerization provides the molecular basis for the Syt1 clamping function. This follows from the investigation of a designed mutation (F349A), which selectively destabilizes Syt1 oligomerization. Using a combination of fluorescence imaging and electrophysiology in neocortical synapses, we show that Syt1F349A is more efficient than wild-type Syt1 (Syt1WT) in triggering synchronous transmitter release but fails to clamp spontaneous and synaptotagmin 7 (Syt7)-mediated asynchronous release components both in rescue (Syt1−/− knockout background) and dominant-interference (Syt1+/+ background) conditions. Thus, we conclude that Ca2+-sensitive Syt1 oligomers, acting as an exocytosis clamp, are critical for maintaining the balance among the different modes of neurotransmitter release.
Collapse
|
29
|
Synaptotagmin-1 and Doc2b Exhibit Distinct Membrane-Remodeling Mechanisms. Biophys J 2019; 118:643-656. [PMID: 31952804 DOI: 10.1016/j.bpj.2019.12.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 12/13/2019] [Accepted: 12/16/2019] [Indexed: 11/24/2022] Open
Abstract
Synaptotagmin-1 (Syt1) is a calcium sensor protein that is critical for neurotransmission and is therefore extensively studied. Here, we use pairs of optically trapped beads coated with SNARE-free synthetic membranes to investigate Syt1-induced membrane remodeling. This activity is compared with that of Doc2b, which contains a conserved C2AB domain and induces membrane tethering and hemifusion in this cell-free model. We find that the soluble C2AB domain of Syt1 strongly affects the probability and strength of membrane-membrane interactions in a strictly Ca2+- and protein-dependent manner. Single-membrane loading of Syt1 yielded the highest probability and force of membrane interactions, whereas in contrast, Doc2b was more effective after loading both membranes. A lipid-mixing assay with confocal imaging reveals that both Syt1 and Doc2b are able to induce hemifusion; however, significantly higher Syt1 concentrations are required. Consistently, both C2AB fragments cause a reduction in the membrane-bending modulus, as measured by a method based on atomic force microscopy. This lowering of the energy required for membrane deformation may contribute to Ca2+-induced fusion.
Collapse
|
30
|
Varga K, Jiang ZJ, Gong LW. Phosphatidylserine is critical for vesicle fission during clathrin-mediated endocytosis. J Neurochem 2019; 152:48-60. [PMID: 31587282 DOI: 10.1111/jnc.14886] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/25/2019] [Accepted: 10/01/2019] [Indexed: 12/11/2022]
Abstract
Phosphatidylserine (PS), a negatively charged phospholipid present predominantly at the inner leaflet of the plasma membrane, has been widely implicated in many cellular processes including membrane trafficking. Along this line, PS has been demonstrated to be important for endocytosis, however, the involved mechanisms remain uncertain. By monitoring clathrin-mediated endocytosis (CME) of single vesicles in mouse chromaffin cells using cell-attached capacitance measurements that offer millisecond time resolution, we demonstrate in the present study that the fission-pore duration is reduced by PS addition, indicating a stimulatory role of PS in regulating the dynamics of vesicle fission during CME. Furthermore, our results show that the PS-mediated effect on the fission-pore duration is Ca2+ -dependent and abolished in the absence of synaptotagmin 1 (Syt1), implying that Syt1 is necessary for the stimulatory role of PS in vesicle fission during CME. Consistently, a Syt1 mutant with a defective PS-Syt1 interaction increases the fission-pore duration. Taken together, our study suggests that PS-Syt1 interaction may be critical in regulating fission dynamics during CME.
Collapse
Affiliation(s)
- Kelly Varga
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA.,Department of Biological Sciences, University of North Texas at Dallas, Dallas, Texas, USA
| | - Zhong-Jiao Jiang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Liang-Wei Gong
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
31
|
Grushin K, Wang J, Coleman J, Rothman JE, Sindelar CV, Krishnakumar SS. Structural basis for the clamping and Ca 2+ activation of SNARE-mediated fusion by synaptotagmin. Nat Commun 2019; 10:2413. [PMID: 31160571 PMCID: PMC6546687 DOI: 10.1038/s41467-019-10391-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 05/08/2019] [Indexed: 12/22/2022] Open
Abstract
Synapotagmin-1 (Syt1) interacts with both SNARE proteins and lipid membranes to synchronize neurotransmitter release to calcium (Ca2+) influx. Here we report the cryo-electron microscopy structure of the Syt1-SNARE complex on anionic-lipid containing membranes. Under resting conditions, the Syt1 C2 domains bind the membrane with a magnesium (Mg2+)-mediated partial insertion of the aliphatic loops, alongside weak interactions with the anionic lipid headgroups. The C2B domain concurrently interacts the SNARE bundle via the 'primary' interface and is positioned between the SNAREpins and the membrane. In this configuration, Syt1 is projected to sterically delay the complete assembly of the associated SNAREpins and thus, contribute to clamping fusion. This Syt1-SNARE organization is disrupted upon Ca2+-influx as Syt1 reorients into the membrane, likely displacing the attached SNAREpins and reversing the fusion clamp. We thus conclude that the cation (Mg2+/Ca2+) dependent membrane interaction is a key determinant of the dual clamp/activator function of Synaptotagmin-1.
Collapse
Affiliation(s)
- Kirill Grushin
- Department of Cell Biology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Jing Wang
- Department of Cell Biology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Jeff Coleman
- Department of Cell Biology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - James E Rothman
- Department of Cell Biology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Charles V Sindelar
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
| | - Shyam S Krishnakumar
- Department of Cell Biology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, Queens Square House, London, WC1 3BG, UK.
| |
Collapse
|
32
|
Fenske P, Grauel MK, Brockmann MM, Dorrn AL, Trimbuch T, Rosenmund C. Autaptic cultures of human induced neurons as a versatile platform for studying synaptic function and neuronal morphology. Sci Rep 2019; 9:4890. [PMID: 30894602 PMCID: PMC6427022 DOI: 10.1038/s41598-019-41259-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 03/05/2019] [Indexed: 02/08/2023] Open
Abstract
Recently developed technology to differentiate induced pluripotent stem cells (iPSCs) into human induced neurons (iNs) provides an exciting opportunity to study the function of human neurons. However, functional characterisations of iNs have been hampered by the reliance on mass culturing protocols which do not allow assessment of synaptic release characteristics and neuronal morphology at the individual cell level with quantitative precision. Here, we have developed for the first time a protocol to generate autaptic cultures of iPSC-derived iNs. We show that our method efficiently generates mature, autaptic iNs with robust spontaneous and action potential-driven synaptic transmission. The synaptic responses are sensitive to modulation by metabotropic receptor agonists as well as potentiation by acute phorbol ester application. Finally, we demonstrate loss of evoked and spontaneous release by Unc13A knockdown. This culture system provides a versatile platform allowing for quantitative and integrative assessment of morphophysiological and molecular parameters underlying human synaptic transmission.
Collapse
Affiliation(s)
- Pascal Fenske
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin, 10117, Berlin, Germany
| | - M Katharina Grauel
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin, 10117, Berlin, Germany
| | - Marisa M Brockmann
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin, 10117, Berlin, Germany
| | - Anja L Dorrn
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin, 10117, Berlin, Germany
| | - Thorsten Trimbuch
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin, 10117, Berlin, Germany
| | - Christian Rosenmund
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117, Berlin, Germany. .,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin, 10117, Berlin, Germany. .,Berlin Institute of Health, Anna-Louise-Karsch-Straße 2, 10178, Berlin, Germany.
| |
Collapse
|
33
|
Quade B, Camacho M, Zhao X, Orlando M, Trimbuch T, Xu J, Li W, Nicastro D, Rosenmund C, Rizo J. Membrane bridging by Munc13-1 is crucial for neurotransmitter release. eLife 2019; 8:42806. [PMID: 30816091 PMCID: PMC6407922 DOI: 10.7554/elife.42806] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 02/22/2019] [Indexed: 11/13/2022] Open
Abstract
Munc13-1 plays a crucial role in neurotransmitter release. We recently proposed that the C-terminal region encompassing the C1, C2B, MUN and C2C domains of Munc13-1 (C1C2BMUNC2C) bridges the synaptic vesicle and plasma membranes through interactions involving the C2C domain and the C1-C2B region. However, the physiological relevance of this model has not been demonstrated. Here we show that C1C2BMUNC2C bridges membranes through opposite ends of its elongated structure. Mutations in putative membrane-binding sites of the C2C domain disrupt the ability of C1C2BMUNC2C to bridge liposomes and to mediate liposome fusion in vitro. These mutations lead to corresponding disruptive effects on synaptic vesicle docking, priming, and Ca2+-triggered neurotransmitter release in mouse neurons. Remarkably, these effects include an almost complete abrogation of release by a single residue substitution in this 200 kDa protein. These results show that bridging the synaptic vesicle and plasma membranes is a central function of Munc13-1.
Collapse
Affiliation(s)
- 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
| | - Marcial Camacho
- Institut für Neurophysiologie, Charité - Universitätsmedizin, Berlin, Germany.,NeuroCure Cluster of Excellence, Berlin, Germany
| | - Xiaowei Zhao
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Marta Orlando
- Institut für Neurophysiologie, Charité - Universitätsmedizin, Berlin, Germany.,NeuroCure Cluster of Excellence, Berlin, Germany
| | - Thorsten Trimbuch
- Institut für Neurophysiologie, Charité - Universitätsmedizin, Berlin, Germany.,NeuroCure Cluster of Excellence, Berlin, Germany
| | - 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
| | - Wei Li
- 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
| | - Daniela Nicastro
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Christian Rosenmund
- Institut für Neurophysiologie, Charité - Universitätsmedizin, Berlin, Germany.,NeuroCure Cluster of Excellence, Berlin, Germany
| | - 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
|
34
|
Biochemical studies of membrane fusion at the single-particle level. Prog Lipid Res 2019; 73:92-100. [PMID: 30611882 DOI: 10.1016/j.plipres.2019.01.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 01/01/2019] [Accepted: 01/02/2019] [Indexed: 01/21/2023]
Abstract
To study membrane fusion mediated by synaptic proteins, proteoliposomes have been widely used for in vitro ensemble measurements with limited insights into the fusion mechanism. Single-particle techniques have proven to be powerful in overcoming the limitations of traditional ensemble methods. Here, we summarize current single-particle methods in biophysical and biochemical studies of fusion mediated by soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) and other synaptic proteins, together with their advantages and limitations.
Collapse
|
35
|
Bello OD, Jouannot O, Chaudhuri A, Stroeva E, Coleman J, Volynski KE, Rothman JE, Krishnakumar SS. Synaptotagmin oligomerization is essential for calcium control of regulated exocytosis. Proc Natl Acad Sci U S A 2018; 115:E7624-E7631. [PMID: 30038018 PMCID: PMC6094142 DOI: 10.1073/pnas.1808792115] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Regulated exocytosis, which underlies many intercellular signaling events, is a tightly controlled process often triggered by calcium ion(s) (Ca2+). Despite considerable insight into the central components involved, namely, the core fusion machinery [soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE)] and the principal Ca2+ sensor [C2-domain proteins like synaptotagmin (Syt)], the molecular mechanism of Ca2+-dependent release has been unclear. Here, we report that the Ca2+-sensitive oligomers of Syt1, a conserved structural feature among several C2-domain proteins, play a critical role in orchestrating Ca2+-coupled vesicular release. This follows from pHluorin-based imaging of single-vesicle exocytosis in pheochromocytoma (PC12) cells showing that selective disruption of Syt1 oligomerization using a structure-directed mutation (F349A) dramatically increases the normally low levels of constitutive exocytosis to effectively occlude Ca2+-stimulated release. We propose a parsimonious model whereby Ca2+-sensitive oligomers of Syt (or a similar C2-domain protein) assembled at the site of docking physically block spontaneous fusion until disrupted by Ca2+ Our data further suggest Ca2+-coupled vesicular release is triggered by removal of the inhibition, rather than by direct activation of the fusion machinery.
Collapse
Affiliation(s)
- Oscar D Bello
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, WC1N 3BG London, United Kingdom
| | - Ouardane Jouannot
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520
| | - Arunima Chaudhuri
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520
| | - Ekaterina Stroeva
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520
| | - Jeff Coleman
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520
| | - Kirill E Volynski
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, WC1N 3BG London, United Kingdom
| | - James E Rothman
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520;
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, WC1N 3BG London, United Kingdom
| | - Shyam S Krishnakumar
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520;
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, WC1N 3BG London, United Kingdom
| |
Collapse
|
36
|
Brunger AT, Leitz J, Zhou Q, Choi UB, Lai Y. Ca 2+-Triggered Synaptic Vesicle Fusion Initiated by Release of Inhibition. Trends Cell Biol 2018; 28:631-645. [PMID: 29706534 PMCID: PMC6056330 DOI: 10.1016/j.tcb.2018.03.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/17/2018] [Accepted: 03/26/2018] [Indexed: 12/20/2022]
Abstract
Recent structural and functional studies of the synaptic vesicle fusion machinery suggest an inhibited tripartite complex consisting of neuronal soluble N-ethylmaleimide sensitive factor attachment protein receptors (SNAREs), synaptotagmin, and complexin prior to Ca2+-triggered synaptic vesicle fusion. We speculate that Ca2+-triggered fusion commences with the release of inhibition by Ca2+ binding to synaptotagmin C2 domains. Subsequently, fusion is assisted by SNARE complex zippering and by active membrane remodeling properties of synaptotagmin. This additional, inhibitory role of synaptotagmin may be a general principle since other recent studies suggest that Ca2+ binding to extended synaptotagmin C2 domains enables lipid transport by releasing an inhibited state of the system, and that Munc13 may nominally be in an inhibited state, which is released upon Ca2+ binding to one of its C2 domains.
Collapse
Affiliation(s)
- Axel T Brunger
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA; Department of Structural Biology, Stanford University, Stanford, CA, USA; Department of Photon Science, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| | - Jeremy Leitz
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA; Department of Structural Biology, Stanford University, Stanford, CA, USA; Department of Photon Science, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Qiangjun Zhou
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA; Department of Structural Biology, Stanford University, Stanford, CA, USA; Department of Photon Science, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Ucheor B Choi
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA; Department of Structural Biology, Stanford University, Stanford, CA, USA; Department of Photon Science, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Ying Lai
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA; Department of Structural Biology, Stanford University, Stanford, CA, USA; Department of Photon Science, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
37
|
Synergistic control of neurotransmitter release by different members of the synaptotagmin family. Curr Opin Neurobiol 2018; 51:154-162. [DOI: 10.1016/j.conb.2018.05.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/19/2018] [Accepted: 05/21/2018] [Indexed: 01/06/2023]
|
38
|
Park Y, Ryu JK. Models of synaptotagmin-1 to trigger Ca 2+ -dependent vesicle fusion. FEBS Lett 2018; 592:3480-3492. [PMID: 30004579 DOI: 10.1002/1873-3468.13193] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/02/2018] [Accepted: 07/06/2018] [Indexed: 11/08/2022]
Abstract
Vesicles in neurons and neuroendocrine cells store neurotransmitters and peptide hormones, which are released by vesicle fusion in response to Ca2+ -evoking stimuli. Synaptotagmin-1 (Syt1), a Ca2+ sensor, mediates ultrafast exocytosis in neurons and neuroendocrine cells. After vesicle docking, Syt1 has two main groups of binding partners: anionic phospholipids and the SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptors) complex. The molecular mechanisms by which Syt1 triggers vesicle fusion remain controversial. This Review introduces and summarizes six molecular models of Syt1: (a) Syt1 triggers SNARE unclamping by displacing complexin, (b) Syt1 clamps SNARE zippering, (c) Syt1 causes membrane curvature, (d) membrane bridging by Syt1, (e) Syt1 is a vesicle-plasma membrane distance regulator, and (f) Syt1 undergoes circular oligomerization. We discuss important conditions to test Syt1 activity in vitro and attempt to illustrate the possible roles of Syt1.
Collapse
Affiliation(s)
- Yongsoo Park
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | - Je-Kyung Ryu
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, The Netherlands
| |
Collapse
|
39
|
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: 134] [Impact Index Per Article: 22.3] [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
|
40
|
MacDougall DD, Lin Z, Chon NL, Jackman SL, Lin H, Knight JD, Anantharam A. The high-affinity calcium sensor synaptotagmin-7 serves multiple roles in regulated exocytosis. J Gen Physiol 2018; 150:783-807. [PMID: 29794152 PMCID: PMC5987875 DOI: 10.1085/jgp.201711944] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 05/07/2018] [Indexed: 12/19/2022] Open
Abstract
MacDougall et al. review the structure and function of the calcium sensor synaptotagmin-7 in exocytosis. Synaptotagmin (Syt) proteins comprise a 17-member family, many of which trigger exocytosis in response to calcium. Historically, most studies have focused on the isoform Syt-1, which serves as the primary calcium sensor in synchronous neurotransmitter release. Recently, Syt-7 has become a topic of broad interest because of its extreme calcium sensitivity and diversity of roles in a wide range of cell types. Here, we review the known and emerging roles of Syt-7 in various contexts and stress the importance of its actions. Unique functions of Syt-7 are discussed in light of recent imaging, electrophysiological, and computational studies. Particular emphasis is placed on Syt-7–dependent regulation of synaptic transmission and neuroendocrine cell secretion. Finally, based on biochemical and structural data, we propose a mechanism to link Syt-7’s role in membrane fusion with its role in subsequent fusion pore expansion via strong calcium-dependent phospholipid binding.
Collapse
Affiliation(s)
| | - Zesen Lin
- Department of Pharmacology, University of Michigan, Ann Arbor, MI
| | - Nara L Chon
- Department of Chemistry, University of Colorado, Denver, CO
| | - Skyler L Jackman
- Vollum Institute, Oregon Health & Science University, Portland, OR
| | - Hai Lin
- Department of Chemistry, University of Colorado, Denver, CO
| | | | - Arun Anantharam
- Department of Pharmacology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
41
|
Kaempf N, Maritzen T. Safeguards of Neurotransmission: Endocytic Adaptors as Regulators of Synaptic Vesicle Composition and Function. Front Cell Neurosci 2017; 11:320. [PMID: 29085282 PMCID: PMC5649181 DOI: 10.3389/fncel.2017.00320] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 09/26/2017] [Indexed: 11/13/2022] Open
Abstract
Communication between neurons relies on neurotransmitters which are released from synaptic vesicles (SVs) upon Ca2+ stimuli. To efficiently load neurotransmitters, sense the rise in intracellular Ca2+ and fuse with the presynaptic membrane, SVs need to be equipped with a stringently controlled set of transmembrane proteins. In fact, changes in SV protein composition quickly compromise neurotransmission and most prominently give rise to epileptic seizures. During exocytosis SVs fully collapse into the presynaptic membrane and consequently have to be replenished to sustain neurotransmission. Therefore, surface-stranded SV proteins have to be efficiently retrieved post-fusion to be used for the generation of a new set of fully functional SVs, a process in which dedicated endocytic sorting adaptors play a crucial role. The question of how the precise reformation of SVs is achieved is intimately linked to how SV membranes are retrieved. For a long time both processes were believed to be two sides of the same coin since Clathrin-mediated endocytosis (CME), the proposed predominant SV recycling mode, will jointly retrieve SV membranes and proteins. However, with the recent proposal of Clathrin-independent SV recycling pathways SV membrane retrieval and SV reformation turn into separable events. This review highlights the progress made in unraveling the molecular mechanisms mediating the high-fidelity retrieval of SV proteins and discusses how the gathered knowledge about SV protein recycling fits in with the new notions of SV membrane endocytosis.
Collapse
Affiliation(s)
- Natalie Kaempf
- Molecular Physiology and Cell Biology Section, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Tanja Maritzen
- Molecular Physiology and Cell Biology Section, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| |
Collapse
|
42
|
Wickner W, Rizo J. A cascade of multiple proteins and lipids catalyzes membrane fusion. Mol Biol Cell 2017; 28:707-711. [PMID: 28292915 PMCID: PMC5349777 DOI: 10.1091/mbc.e16-07-0517] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 01/05/2017] [Accepted: 01/10/2017] [Indexed: 11/11/2022] Open
Abstract
Recent studies suggest revisions to the SNARE paradigm of membrane fusion. Membrane tethers and/or SNAREs recruit proteins of the Sec 1/Munc18 family to catalyze SNARE assembly into trans-complexes. SNARE-domain zippering draws the bilayers into immediate apposition and provides a platform to position fusion triggers such as Sec 17/α-SNAP and/or synaptotagmin, which insert their apolar "wedge" domains into the bilayers, initiating the lipid rearrangements of fusion.
Collapse
Affiliation(s)
- William Wickner
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755 )
| | - Josep Rizo
- Departments of Biophysics, Biochemistry, and Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390 )
| |
Collapse
|
43
|
Exceptionally tight membrane-binding may explain the key role of the synaptotagmin-7 C 2A domain in asynchronous neurotransmitter release. Proc Natl Acad Sci U S A 2017; 114:E8518-E8527. [PMID: 28923929 DOI: 10.1073/pnas.1710708114] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Synaptotagmins (Syts) act as Ca2+ sensors in neurotransmitter release by virtue of Ca2+-binding to their two C2 domains, but their mechanisms of action remain unclear. Puzzlingly, Ca2+-binding to the C2B domain appears to dominate Syt1 function in synchronous release, whereas Ca2+-binding to the C2A domain mediates Syt7 function in asynchronous release. Here we show that crystal structures of the Syt7 C2A domain and C2AB region, and analyses of intrinsic Ca2+-binding to the Syt7 C2 domains using isothermal titration calorimetry, did not reveal major differences that could explain functional differentiation between Syt7 and Syt1. However, using liposome titrations under Ca2+ saturating conditions, we show that the Syt7 C2A domain has a very high membrane affinity and dominates phospholipid binding to Syt7 in the presence or absence of l-α-phosphatidylinositol 4,5-diphosphate (PIP2). For Syt1, the two Ca2+-saturated C2 domains have similar affinities for membranes lacking PIP2, but the C2B domain dominates binding to PIP2-containing membranes. Mutagenesis revealed that the dramatic differences in membrane affinity between the Syt1 and Syt7 C2A domains arise in part from apparently conservative residue substitutions, showing how striking biochemical and functional differences can result from the cumulative effects of subtle residue substitutions. Viewed together, our results suggest that membrane affinity may be a key determinant of the functions of Syt C2 domains in neurotransmitter release.
Collapse
|
44
|
Wang J, Li F, Bello OD, Sindelar CV, Pincet F, Krishnakumar SS, Rothman JE. Circular oligomerization is an intrinsic property of synaptotagmin. eLife 2017; 6. [PMID: 28850328 PMCID: PMC5576491 DOI: 10.7554/elife.27441] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 08/11/2017] [Indexed: 11/13/2022] Open
Abstract
Previously, we showed that synaptotagmin1 (Syt1) forms Ca2+-sensitive ring-like oligomers on membranes containing acidic lipids and proposed a potential role in regulating neurotransmitter release (Zanetti et al., 2016). Here, we report that Syt1 assembles into similar ring-like oligomers in solution when triggered by naturally occurring polyphosphates (PIP2 and ATP) and magnesium ions (Mg2+). These soluble Syt1 rings were observed by electron microscopy and independently demonstrated and quantified using fluorescence correlation spectroscopy. Oligomerization is triggered when polyphosphates bind to the polylysine patch in C2B domain and is stabilized by Mg2+, which neutralizes the Ca2+-binding aspartic acids that likely contribute to the C2B interface in the oligomer. Overall, our data show that ring-like polymerization is an intrinsic property of Syt1 with reasonable affinity that can be triggered by the vesicle docking C2B-PIP2 interaction and raise the possibility that Syt1 rings could pre-form on the synaptic vesicle to facilitate docking.
Collapse
Affiliation(s)
- Jing Wang
- Departments of Cell Biology, Yale University School of Medicine, New Haven, United States
| | - Feng Li
- Departments of Cell Biology, Yale University School of Medicine, New Haven, United States
| | - Oscar D Bello
- Departments of Cell Biology, Yale University School of Medicine, New Haven, United States.,Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, London, United Kingdom
| | - Charles Vaughn Sindelar
- Departments of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, United States
| | - Frédéric Pincet
- Departments of Cell Biology, Yale University School of Medicine, New Haven, United States.,Laboratoire de Physique Statistique, UMR CNRS 8550 Associée aux Universités Paris 6 et Paris 7, Paris, France
| | - Shyam S Krishnakumar
- Departments of Cell Biology, Yale University School of Medicine, New Haven, United States.,Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, London, United Kingdom
| | - James E Rothman
- Departments of Cell Biology, Yale University School of Medicine, New Haven, United States.,Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
45
|
Lipstein N, Verhoeven-Duif NM, Michelassi FE, Calloway N, van Hasselt PM, Pienkowska K, van Haaften G, van Haelst MM, van Empelen R, Cuppen I, van Teeseling HC, Evelein AMV, Vorstman JA, Thoms S, Jahn O, Duran KJ, Monroe GR, Ryan TA, Taschenberger H, Dittman JS, Rhee JS, Visser G, Jans JJ, Brose N. Synaptic UNC13A protein variant causes increased neurotransmission and dyskinetic movement disorder. J Clin Invest 2017; 127:1005-1018. [PMID: 28192369 DOI: 10.1172/jci90259] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 12/15/2016] [Indexed: 12/13/2022] Open
Abstract
Munc13 proteins are essential regulators of neurotransmitter release at nerve cell synapses. They mediate the priming step that renders synaptic vesicles fusion-competent, and their genetic elimination causes a complete block of synaptic transmission. Here we have described a patient displaying a disorder characterized by a dyskinetic movement disorder, developmental delay, and autism. Using whole-exome sequencing, we have shown that this condition is associated with a rare, de novo Pro814Leu variant in the major human Munc13 paralog UNC13A (also known as Munc13-1). Electrophysiological studies in murine neuronal cultures and functional analyses in Caenorhabditis elegans revealed that the UNC13A variant causes a distinct dominant gain of function that is characterized by increased fusion propensity of synaptic vesicles, which leads to increased initial synaptic vesicle release probability and abnormal short-term synaptic plasticity. Our study underscores the critical importance of fine-tuned presynaptic control in normal brain function. Further, it adds the neuronal Munc13 proteins and the synaptic vesicle priming process that they control to the known etiological mechanisms of psychiatric and neurological synaptopathies.
Collapse
|
46
|
Pinheiro PS, Houy S, Sørensen JB. C2-domain containing calcium sensors in neuroendocrine secretion. J Neurochem 2016; 139:943-958. [DOI: 10.1111/jnc.13865] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 09/17/2016] [Accepted: 10/05/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Paulo S. Pinheiro
- Center for Neuroscience and Cell Biology; University of Coimbra; Coimbra Portugal
| | - Sébastien Houy
- Department of Neuroscience and Pharmacology; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - Jakob B. Sørensen
- Department of Neuroscience and Pharmacology; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| |
Collapse
|
47
|
Qiu X, Ge J, Gao Y, Teng M, Niu L. Structural analysis of Ca 2+-binding pocket of synaptotagmin 5 C2A domain. Int J Biol Macromol 2016; 95:946-953. [PMID: 27793683 DOI: 10.1016/j.ijbiomac.2016.10.083] [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: 04/09/2015] [Revised: 10/09/2016] [Accepted: 10/24/2016] [Indexed: 10/20/2022]
Abstract
Synaptotagmins constitute a family of multifunctional integral membrane proteins found predominantly on vesicles in neural and endocrine tissues. 17 isoforms of synaptotagmin family in mammals have been identified, 7 isoforms among them are known to be able to bind Ca2+ via their C2 domains. This study presents the crystal structure of the first C2 domain (C2A domain) of synaptotagmin 5 complexed with Ca2+ at 1.90Å resolution. Comparison of the Ca2+-binding pocket of synaptotagmin 5 C2A domain with other synaptotagmin C2 domains demonstrated that a serine residue locating at Ca2+-binding loop probably responsible to the conformational variation of Ca2+-binding pocket, and thus impacts the Ca2+-binding mechanism of C2 domain, which is verified by structural analysis of the serine mutant and Ca2+-binding assays via isothermal titration calorimetry. Alteration of Ca2+-binding mechanism might be correlated with different Ca2+ response rates of synaptotagmins, which is the basis of the functions of synaptotagmins in regulating various types of Ca2+-triggered vesicle-membrane fusion processes.
Collapse
Affiliation(s)
- Xiaoting Qiu
- Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, Zhejiang 315211, PR China; School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315211, PR China; Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, PR China; Key Laboratory of Structural Biology, Chinese Academy of Sciences, Hefei, Anhui 230026, PR China
| | - Junyi Ge
- Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, Zhejiang 315211, PR China; School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Yongxiang Gao
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, PR China; Key Laboratory of Structural Biology, Chinese Academy of Sciences, Hefei, Anhui 230026, PR China
| | - Maikun Teng
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, PR China; Key Laboratory of Structural Biology, Chinese Academy of Sciences, Hefei, Anhui 230026, PR China.
| | - Liwen Niu
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, PR China; Key Laboratory of Structural Biology, Chinese Academy of Sciences, Hefei, Anhui 230026, PR China.
| |
Collapse
|
48
|
Pérez-Lara Á, Thapa A, Nyenhuis SB, Nyenhuis DA, Halder P, Tietzel M, Tittmann K, Cafiso DS, Jahn R. PtdInsP 2 and PtdSer cooperate to trap synaptotagmin-1 to the plasma membrane in the presence of calcium. eLife 2016; 5. [PMID: 27791979 PMCID: PMC5123861 DOI: 10.7554/elife.15886] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 10/25/2016] [Indexed: 12/16/2022] Open
Abstract
The Ca2+-sensor synaptotagmin-1 that triggers neuronal exocytosis binds to negatively charged membrane lipids (mainly phosphatidylserine (PtdSer) and phosphoinositides (PtdIns)) but the molecular details of this process are not fully understood. Using quantitative thermodynamic, kinetic and structural methods, we show that synaptotagmin-1 (from Rattus norvegicus and expressed in Escherichia coli) binds to PtdIns(4,5)P2 via a polybasic lysine patch in the C2B domain, which may promote the priming or docking of synaptic vesicles. Ca2+ neutralizes the negative charges of the Ca2+-binding sites, resulting in the penetration of synaptotagmin-1 into the membrane, via binding of PtdSer, and an increase in the affinity of the polybasic lysine patch to phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P2). These Ca2+-induced events decrease the dissociation rate of synaptotagmin-1 membrane binding while the association rate remains unchanged. We conclude that both membrane penetration and the increased residence time of synaptotagmin-1 at the plasma membrane are crucial for triggering exocytotic membrane fusion. DOI:http://dx.doi.org/10.7554/eLife.15886.001 The human nervous system contains billions of neurons that communicate with each other across junctions called synapses. When a neuron is activated, the levels of calcium ions inside the cell rise. This causes molecules called neurotransmitters to be released from the neuron at a synapse to make contact with the second neuron. The neurotransmitters are stored inside cells within compartments known as synaptic vesicles and are released when these vesicles fuse with the membrane surrounding the cell. Proteins called SNAREs regulate the membrane fusion process. These proteins assemble into bundles that help to drive vesicle and cell membranes together. Another protein called synaptotagmin-1 sticks out from the vesicle membrane and senses the levels of calcium ions in the cell to trigger membrane fusion at the right time. Synaptotagmin-1 has two regions that can bind to calcium ions, known as the C2 domains. When calcium ion levels rise, these domains insert into the cell membrane by binding to two fat molecules in the membrane called phosphatidylserine (PtdSer) and phosphatidylinositol 4,5-bisphosphate (PtdInsP2). Synaptotagmin-1 also interacts with the SNARE proteins, but it is not known whether synaptotagmin-1 triggers fusion by binding directly to SNAREs, or by the way it inserts into the cell membrane. Pérez-Lara et al. used several biophysical methods to investigate how synaptotagmin-1 binds to PtdSer and PtdInsP2. The experiments show that these molecules bind to different regions of synaptotagmin-1 and work together to attach the protein to the cell membrane and insert the C2 domains. Calcium ions increase the affinity of synaptotagmin-1 binding to the cell membrane by making it harder for synaptotagmin-1 to separate from the membrane, rather than by increasing its ability to bind to it. Further experiments show that synaptotagmin-1 prefers to bind to membranes that contain PtdInsP2 over binding to the SNARE proteins. Together, the findings of Pérez-Lara et al. suggest that calcium ions may trigger the release of neurotransmitters by trapping synaptotagmin-1 at the cell membrane rather than by directly affecting how it interacts with SNARE proteins. Further work will be needed to establish exactly how the SNARE proteins, PtdInsP2 and synaptotagmin-1 interact. DOI:http://dx.doi.org/10.7554/eLife.15886.002
Collapse
Affiliation(s)
- Ángel Pérez-Lara
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Anusa Thapa
- Department of Chemistry and Center for Membrane Biology, University of Virginia, Charlottesville, United States
| | - Sarah B Nyenhuis
- Department of Chemistry and Center for Membrane Biology, University of Virginia, Charlottesville, United States
| | - David A Nyenhuis
- Department of Chemistry and Center for Membrane Biology, University of Virginia, Charlottesville, United States
| | - Partho Halder
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Michael Tietzel
- Department of Molecular Enzymology, Göttingen Center for Molecular Biosciences, Georg-August University Göttingen, Göttingen, Germany
| | - Kai Tittmann
- Department of Molecular Enzymology, Göttingen Center for Molecular Biosciences, Georg-August University Göttingen, Göttingen, Germany
| | - David S Cafiso
- Department of Chemistry and Center for Membrane Biology, University of Virginia, Charlottesville, United States
| | - Reinhard Jahn
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| |
Collapse
|
49
|
Zanetti MN, Bello OD, Wang J, Coleman J, Cai Y, Sindelar CV, Rothman JE, Krishnakumar SS. Ring-like oligomers of Synaptotagmins and related C2 domain proteins. eLife 2016; 5. [PMID: 27434670 PMCID: PMC4977156 DOI: 10.7554/elife.17262] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/18/2016] [Indexed: 01/25/2023] Open
Abstract
We recently reported that the C2AB portion of Synaptotagmin 1 (Syt1) could self-assemble into Ca(2+)-sensitive ring-like oligomers on membranes, which could potentially regulate neurotransmitter release. Here we report that analogous ring-like oligomers assemble from the C2AB domains of other Syt isoforms (Syt2, Syt7, Syt9) as well as related C2 domain containing protein, Doc2B and extended Synaptotagmins (E-Syts). Evidently, circular oligomerization is a general and conserved structural aspect of many C2 domain proteins, including Synaptotagmins. Further, using electron microscopy combined with targeted mutations, we show that under physiologically relevant conditions, both the Syt1 ring assembly and its rapid disruption by Ca(2+) involve the well-established functional surfaces on the C2B domain that are important for synaptic transmission. Our data suggests that ring formation may be triggered at an early step in synaptic vesicle docking and positions Syt1 to synchronize neurotransmitter release to Ca(2+) influx.
Collapse
Affiliation(s)
- Maria N Zanetti
- Department of Cell Biology, Yale School of Medicine, New Haven, United States
| | - Oscar D Bello
- Department of Cell Biology, Yale School of Medicine, New Haven, United States
| | - Jing Wang
- Department of Cell Biology, Yale School of Medicine, New Haven, United States
| | - Jeff Coleman
- Department of Cell Biology, Yale School of Medicine, New Haven, United States
| | - Yiying Cai
- Department of Cell Biology, Yale School of Medicine, New Haven, United States
| | - Charles V Sindelar
- Department of Molecular Biophysics and Biochemistry, Yale School of Medicine, New Haven, United States
| | - James E Rothman
- Department of Cell Biology, Yale School of Medicine, New Haven, United States
| | | |
Collapse
|
50
|
Cork KM, Van Hook MJ, Thoreson WB. Mechanisms, pools, and sites of spontaneous vesicle release at synapses of rod and cone photoreceptors. Eur J Neurosci 2016; 44:2015-27. [PMID: 27255664 DOI: 10.1111/ejn.13288] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 05/21/2016] [Accepted: 05/24/2016] [Indexed: 01/31/2023]
Abstract
Photoreceptors have depolarized resting potentials that stimulate calcium-dependent release continuously from a large vesicle pool but neurons can also release vesicles without stimulation. We characterized the Ca(2+) dependence, vesicle pools, and release sites involved in spontaneous release at photoreceptor ribbon synapses. In whole-cell recordings from light-adapted horizontal cells (HCs) of tiger salamander retina, we detected miniature excitatory post-synaptic currents (mEPSCs) when no stimulation was applied to promote exocytosis. Blocking Ca(2+) influx by lowering extracellular Ca(2+) , by application of Cd(2+) and other agents reduced the frequency of mEPSCs but did not eliminate them, indicating that mEPSCs can occur independently of Ca(2+) . We also measured release presynaptically from rods and cones by examining quantal glutamate transporter anion currents. Presynaptic quantal event frequency was reduced by Cd(2+) or by increased intracellular Ca(2+) buffering in rods, but not in cones, that were voltage clamped at -70 mV. By inhibiting the vesicle cycle with bafilomycin, we found the frequency of mEPSCs declined more rapidly than the amplitude of evoked excitatory post-synaptic currents (EPSCs) suggesting a possible separation between vesicle pools in evoked and spontaneous exocytosis. We mapped sites of Ca(2+) -independent release using total internal reflectance fluorescence (TIRF) microscopy to visualize fusion of individual vesicles loaded with dextran-conjugated pHrodo. Spontaneous release in rods occurred more frequently at non-ribbon sites than evoked release events. The function of Ca(2+) -independent spontaneous release at continuously active photoreceptor synapses remains unclear, but the low frequency of spontaneous quanta limits their impact on noise.
Collapse
Affiliation(s)
- Karlene M Cork
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, 4050 Durham Research Center, University of Nebraska Medical Center, Omaha, NE, 68198-5840, USA.,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Matthew J Van Hook
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, 4050 Durham Research Center, University of Nebraska Medical Center, Omaha, NE, 68198-5840, USA
| | - Wallace B Thoreson
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, 4050 Durham Research Center, University of Nebraska Medical Center, Omaha, NE, 68198-5840, USA.,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|