1
|
Jia T, Yang F, Qin F, He Y, Han F, Zhang C. Identification of Common Brain Protein and Genetic Loci Between Parkinson's Disease and Lewy Body Dementia. CNS Neurosci Ther 2025; 31:e70370. [PMID: 40202048 PMCID: PMC11979625 DOI: 10.1111/cns.70370] [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: 03/25/2024] [Revised: 02/25/2025] [Accepted: 03/17/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Parkinson's disease (PD) and Lewy body dementia (LBD) have many common features, including clinical manifestations, neurochemistry, and pathology, but little is known about their shared brain proteins and genetic factors. METHODS To identify susceptibility-related brain proteins that are shared between PD and LBD patients, proteome-wide association studies (PWASs) were conducted by integrating human brain protein quantitative trait loci (pQTLs) with large-scale genome-wide association studies (GWASs) of both diseases. Subsequently, pleiotropy-informed conditional false discovery rate (pleioFDR) analysis was performed to identify common risk genetic loci between PD and LBD. Finally, the downregulation of these risk genes in different disease states was validated by differential gene expression analysis. RESULTS PWASs identified 12 PD risk proteins and nine LBD risk proteins, among which TMEM175 (zPD = -7.25, PPD = 4.12E-13; zLBD = -6.02, PLBD = 1.75E-09) and DOC2A (zPD = -4.13, PPD = 3.71E-05; zLBD = -3.91, PLBD = 9.08E-05) were shared. PleioFDR analysis revealed that five genetic risk loci mapped to eight genes associated with PD and LBD, including the proteome-wide significant risk gene TMEM175 (ConjFDR = 5.74E-03). Differential expression analysis verified that TMEM175 was significantly downregulated in the midbrains of PD patients (p = 1.19E-02), and further exploration revealed that TMEM175 was also dramatically downregulated in the substantia nigra of PD patients (p = 1.16E-02) and incidental Lewy body disease patients (p = 7.52E-03). Moreover, TMEM175 was significantly downregulated in induced pluripotent stem cell-derived dopaminergic neurons from PD patients (p = 4.60E-02). CONCLUSION Dysregulation of TMEM175 may confer PD and LBD risk and may be partly responsible for their comorbidity. Our results revealed the common genetic risk factors between PD and LBD, which elucidated the shared genetic basis of these diseases.
Collapse
Affiliation(s)
- Tingting Jia
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Department of Gastroenterology and Hepatology and Sichuan University‐University of Oxford Huaxi Joint Centre for Gastrointestinal CancerWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Fuhua Yang
- Department of NephrologyThe Sixth People's Hospital of ChengduChengduSichuanChina
| | - Fengqin Qin
- Department of NeurologyThe 3rd Affiliated Hospital of Chengdu Medical CollegeChengduSichuanChina
| | - Yongji He
- Clinical Trial Center, National Medical Products Administration key Laboratory for Clinical Research and Evaluation of Innovative DrugsWest China Hospital Sichuan UniversityChengduChina
| | - Feng Han
- Department of Emergency MedicineHainan General Hospital, Hainan Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Chengcheng Zhang
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduSichuanChina
| |
Collapse
|
2
|
Boudriot E, Stephan M, Rabe F, Smigielski L, Schmitt A, Falkai P, Ziller MJ, Rossner MJ, Homan P, Papiol S, Raabe FJ. Genetic Analysis of Retinal Cell Types in Neuropsychiatric Disorders. JAMA Psychiatry 2025; 82:285-295. [PMID: 39775833 PMCID: PMC11883512 DOI: 10.1001/jamapsychiatry.2024.4230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/29/2024] [Indexed: 01/11/2025]
Abstract
Importance As an accessible part of the central nervous system, the retina provides a unique window to study pathophysiological mechanisms of brain disorders in humans. Imaging and electrophysiological studies have revealed retinal alterations across several neuropsychiatric and neurological disorders, but it remains largely unclear which specific cell types and biological mechanisms are involved. Objective To determine whether specific retinal cell types are affected by genomic risk for neuropsychiatric and neurological disorders and to explore the mechanisms through which genomic risk converges in these cell types. Design, Setting, and Participants This genetic association study combined findings from genome-wide association studies in schizophrenia, bipolar disorder, major depressive disorder, multiple sclerosis, Parkinson disease, Alzheimer disease, and stroke with retinal single-cell transcriptomic datasets from humans, macaques, and mice. To identify susceptible cell types, Multi-Marker Analysis of Genomic Annotation (MAGMA) cell-type enrichment analyses were applied and subsequent pathway analyses performed. The cellular top hits were translated to the structural level using retinal optical coherence tomography (acquired between 2009 and 2010) and genotyping data in the large population-based UK Biobank cohort study. Data analysis was conducted between 2022 and 2024. Main Outcomes and Measures Cell type-specific enrichment of genetic risk loading for neuropsychiatric and neurological disorder traits in the gene expression profiles of retinal cells. Results Expression profiles of amacrine cells (interneurons within the retina) were robustly enriched in schizophrenia genetic risk across mammalian species and in different developmental stages. This enrichment was primarily driven by genes involved in synapse biology. Moreover, expression profiles of retinal immune cell populations were enriched in multiple sclerosis genetic risk. No consistent cell-type associations were found for bipolar disorder, major depressive disorder, Parkinson disease, Alzheimer disease, or stroke. On the structural level, higher polygenic risk for schizophrenia was associated with thinning of the ganglion cell inner plexiform layer, which contains dendrites and synaptic connections of amacrine cells (B, -0.09; 95% CI, -0.16 to -0.03; P = .007; n = 36 349; mean [SD] age, 57.50 [8.00] years; 19 859 female [54.63%]). Higher polygenic risk for multiple sclerosis was associated with increased thickness of the retinal nerve fiber layer (B, 0.06; 95% CI, 0.02 to 0.10; P = .007; n = 36 371; mean [SD] age, 57.51 [8.00] years; 19 843 female [54.56%]). Conclusions and Relevance This study provides novel insights into the cellular underpinnings of retinal alterations in neuropsychiatric and neurological disorders and highlights the retina as a potential proxy to study synaptic pathology in schizophrenia.
Collapse
Affiliation(s)
- Emanuel Boudriot
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Marius Stephan
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- Systasy Bioscience, Munich, Germany
| | - Finn Rabe
- Department of Adult Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lukasz Smigielski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Andrea Schmitt
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- German Center for Mental Health, Partner Site Munich-Augsburg, Germany
- Laboratory of Neurosciences, Institute of Psychiatry, University of São Paulo, São Paulo, Brazil
| | - Peter Falkai
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- German Center for Mental Health, Partner Site Munich-Augsburg, Germany
| | - Michael J. Ziller
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry, University of Münster, Münster, Germany
- Center for Soft Nanoscience, University of Münster, Münster, Germany
| | - Moritz J. Rossner
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- Systasy Bioscience, Munich, Germany
| | - Philipp Homan
- Department of Adult Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Sergi Papiol
- Max Planck Institute of Psychiatry, Munich, Germany
- Institute of Psychiatric Phenomics and Genomics, Ludwig Maximilian University Munich, Munich, Germany
| | - Florian J. Raabe
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| |
Collapse
|
3
|
Bose D, Bera M, Norman CA, Timofeeva Y, Volynski KE, Krishnakumar SS. Minimal presynaptic protein machinery governing diverse kinetics of calcium-evoked neurotransmitter release. Nat Commun 2024; 15:10741. [PMID: 39738049 PMCID: PMC11685451 DOI: 10.1038/s41467-024-54960-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
Neurotransmitters are released from synaptic vesicles with remarkable precision in response to presynaptic calcium influx but exhibit significant heterogeneity in exocytosis timing and efficacy based on the recent history of activity. This heterogeneity is critical for information transfer in the brain, yet its molecular basis remains poorly understood. Here, we employ a biochemically-defined fusion assay under physiologically relevant conditions to delineate the minimal protein machinery sufficient to account for various modes of calcium-triggered vesicle fusion dynamics. We find that Synaptotagmin-1, Synaptotagmin-7, and Complexin synergistically restrain SNARE complex assembly, thus preserving vesicles in a stably docked state at rest. Upon calcium activation, Synaptotagmin-1 induces rapid vesicle fusion, while Synaptotagmin-7 mediates delayed fusion. Competitive binding of Synaptotagmin-1 and Synaptotagmin-7 to the same SNAREs, coupled with differential rates of calcium-triggered fusion clamp reversal, govern the overall kinetics of vesicular fusion. Under conditions mimicking sustained neuronal activity, the Synaptotagmin-7 fusion clamp is destabilized by the elevated basal calcium concentration, thereby enhancing the synchronous component of fusion. These findings provide a direct demonstration that a small set of proteins is sufficient to account for how nerve terminals adapt and regulate the calcium-evoked neurotransmitter exocytosis process to support their specialized functions in the nervous system.
Collapse
Affiliation(s)
- Dipayan Bose
- Nanobiology Institute, Yale University, West Haven, CT, USA
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, USA
| | - Manindra Bera
- Nanobiology Institute, Yale University, West Haven, CT, USA
- Department of Cell Biology, School of Medicine, Yale University, New Haven, CT, USA
| | - Christopher A Norman
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
- Department of Computer Science, University of Warwick, Coventry, UK
| | - Yulia Timofeeva
- Department of Computer Science, University of Warwick, Coventry, UK
| | - Kirill E Volynski
- Department of Cell Biology, School of Medicine, Yale University, New Haven, CT, USA.
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK.
| | - Shyam S Krishnakumar
- Nanobiology Institute, Yale University, West Haven, CT, USA.
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, USA.
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK.
| |
Collapse
|
4
|
Ostermann PN, Wu Y, Bowler SA, Siddiqui MA, Herrera A, Sidharta M, Ramnarine K, Martínez-Meza S, St. Bernard LA, Nixon DF, Jones RB, Yamashita M, Ndhlovu LC, Zhou T, Evering TH. A Transcriptional Signature of Induced Neurons Differentiates Virologically Suppressed People Living With HIV from People Without HIV. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619617. [PMID: 39484396 PMCID: PMC11526917 DOI: 10.1101/2024.10.22.619617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Neurocognitive impairment is a prevalent and important co-morbidity in virologically suppressed people living with HIV (PLWH), yet the underlying mechanisms remain elusive and treatments lacking. Here, we explored for the first time, use of participant-derived directly induced neurons (iNs) to model neuronal biology and injury in PLWH. iNs retain age- and disease-related features of the donors, providing unique opportunities to reveal novel aspects of neurological disorders. We obtained primary dermal fibroblasts from six virologically suppressed PLWH (range: 27 - 64 years, median: 53); 83% Male; 50% White) and seven matched people without HIV (PWOH) (range: 27 - 66, median: 55); 71% Male; 57% White). iNs were generated using transcription factors NGN2 and ASCL1, and validated by immunocytochemistry and single-cell-RNAseq. Transcriptomic analysis using bulk-RNAseq identified 29 significantly differentially expressed genes between iNs from PLWH and PWOH. Of these, 16 genes were downregulated and 13 upregulated in PLWH iNs. Protein-protein interaction network mapping indicates that iNs from PLWH exhibit differences in extracellular matrix organization and synaptic transmission. IFI27 was upregulated in iNs from PLWH, which complements independent post-mortem studies demonstrating elevated IFI27 expression in PLWH-derived brain tissue, indicating that iN generation reconstitutes this pathway. Finally, we observed that expression of the FOXL2NB-FOXL2-LINC01391 genome locus is reduced in iNs from PLWH and negatively correlates with neurocognitive impairment. Thus, we have identified an iN gene signature of HIV through direct reprogramming of skin fibroblasts into neurons revealing novel mechanisms of neurocognitive impairment in PLWH.
Collapse
Affiliation(s)
- Philipp N. Ostermann
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Youjun Wu
- The SKI Stem Cell Research Facility, The Center for Stem Cell Biology and Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Scott A. Bowler
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Mohammad Adnan Siddiqui
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY. 10032, USA
| | - Alberto Herrera
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Mega Sidharta
- The SKI Stem Cell Research Facility, The Center for Stem Cell Biology and Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Kiran Ramnarine
- The SKI Stem Cell Research Facility, The Center for Stem Cell Biology and Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Samuel Martínez-Meza
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Leslie Ann St. Bernard
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Douglas F. Nixon
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - R. Brad Jones
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Masahiro Yamashita
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY. 10032, USA
| | - Lishomwa C. Ndhlovu
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ting Zhou
- The SKI Stem Cell Research Facility, The Center for Stem Cell Biology and Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Teresa H. Evering
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
5
|
Esparza D, Lima C, Abuelreich S, Ghaeli I, Hwang J, Oh E, Lenz A, Gu A, Jiang N, Kandeel F, Thurmond DC, Jovanovic-Talisman T. Pancreatic β-cells package double C2-like domain beta protein into extracellular vesicles via tandem C2 domains. Front Endocrinol (Lausanne) 2024; 15:1451279. [PMID: 39497805 PMCID: PMC11532064 DOI: 10.3389/fendo.2024.1451279] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/10/2024] [Indexed: 11/07/2024] Open
Abstract
Introduction Double C2-like domain beta (DOC2B) is a vesicle priming protein critical for glucose-stimulated insulin secretion in β-cells. Individuals with type 1 diabetes (T1D) have lower levels of DOC2B in their residual functional β-cell mass and platelets, a phenotype also observed in a mouse model of T1D. Thus, DOC2B levels could provide important information on β-cell dys(function). Objective Our objective was to evaluate the DOC2B secretome of β-cells. In addition to soluble extracellular protein, we assessed DOC2B localized within membrane-delimited nanoparticles - extracellular vesicles (EVs). Moreover, in rat clonal β-cells, we probed domains required for DOC2B sorting into EVs. Method Using Single Extracellular VEsicle Nanoscopy, we quantified EVs derived from clonal β-cells (human EndoC-βH1, rat INS-1 832/13, and mouse MIN6); two other cell types known to regulate glucose homeostasis and functionally utilize DOC2B (skeletal muscle rat myotube L6-GLUT4myc and human neuronal-like SH-SY5Y cells); and human islets sourced from individuals with no diabetes (ND). EVs derived from ND human plasma, ND human islets, and cell lines were isolated with either size exclusion chromatography or differential centrifugation. Isolated EVs were comprehensively characterized using dotblots, transmission electron microscopy, nanoparticle tracking analysis, and immunoblotting. Results DOC2B was present within EVs derived from ND human plasma, ND human islets, and INS-1 832/13 β-cells. Compared to neuronal-like SH-SY5Y cells and L6-GLUT4myc myotubes, clonal β-cells (EndoC-βH1, INS-1 832/13, and MIN6) produced significantly more EVs. DOC2B levels in EVs (over whole cell lysates) were higher in INS-1 832/13 β-cells compared to L6-GLUT4myc myotubes; SH-SY5Y neuronal-like cells did not release appreciable DOC2B. Mechanistically, we show that DOC2B was localized to the EV lumen; the tandem C2 domains were sufficient to confer sorting to INS-1 832/13 β-cell EVs. Discussion Clonal β-cells and ND human islets produce abundant EVs. In cell culture, appreciable DOC2B can be packaged into EVs, and a small fraction is excreted as a soluble protein. While DOC2B-laden EVs and soluble protein are present in ND plasma, further studies will be necessary to determine if DOC2B originating from β-cells significantly contributes to the plasma secretome.
Collapse
Affiliation(s)
- Diana Esparza
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Carinna Lima
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Sarah Abuelreich
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Ima Ghaeli
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Jinhee Hwang
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Eunjin Oh
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Ayelet Lenz
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Angel Gu
- Department of Translational Research and Cellular Therapeutics, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Nan Jiang
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Fouad Kandeel
- Department of Translational Research and Cellular Therapeutics, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Debbie C. Thurmond
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| | - Tijana Jovanovic-Talisman
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute at City of Hope, Duarte, CA, United States
| |
Collapse
|
6
|
Thoreson WB, Zenisek D. Presynaptic Proteins and Their Roles in Visual Processing by the Retina. Annu Rev Vis Sci 2024; 10:347-375. [PMID: 38621251 PMCID: PMC11536687 DOI: 10.1146/annurev-vision-101322-111204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
The sense of vision begins in the retina, where light is detected and processed through a complex series of synaptic connections into meaningful information relayed to the brain via retinal ganglion cells. Light responses begin as tonic and graded signals in photoreceptors, later emerging from the retina as a series of spikes from ganglion cells. Processing by the retina extracts critical features of the visual world, including spatial frequency, temporal frequency, motion direction, color, contrast, and luminance. To achieve this, the retina has evolved specialized and unique synapse types. These include the ribbon synapses of photoreceptors and bipolar cells, the dendritic synapses of amacrine and horizontal cells, and unconventional synaptic feedback from horizontal cells to photoreceptors. We review these unique synapses in the retina with a focus on the presynaptic molecules and physiological properties that shape their capabilities.
Collapse
Affiliation(s)
- Wallace B Thoreson
- Departments of Ophthalmology & Visual Sciences and Pharmacology & Experimental Neuroscience, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA;
| | - David Zenisek
- Departments of Cellular and Molecular Physiology, Ophthalmology and Visual Sciences, and Neuroscience, Yale University, New Haven, Connecticut, USA;
| |
Collapse
|
7
|
Weingarten DJ, Shrestha A, Orlin DJ, Le Moing CL, Borchardt LA, Jackman SL. Synaptotagmins 3 and 7 mediate the majority of asynchronous release from synapses in the cerebellum and hippocampus. Cell Rep 2024; 43:114595. [PMID: 39116209 PMCID: PMC11410144 DOI: 10.1016/j.celrep.2024.114595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/24/2024] [Accepted: 07/22/2024] [Indexed: 08/10/2024] Open
Abstract
Neurotransmitter release consists of rapid synchronous release followed by longer-lasting asynchronous release (AR). Although the presynaptic proteins that trigger synchronous release are well understood, the mechanisms for AR remain unclear. AR is sustained by low concentrations of intracellular Ca2+ and Sr2+, suggesting the involvement of sensors with high affinities for both ions. Synaptotagmin 7 (SYT7) partly mediates AR, but substantial AR persists in the absence of SYT7. The closely related SYT3 binds Ca2+ and Sr2+ with high affinity, making it a promising candidate to mediate AR. Here, we use knockout mice to study the contribution of SYT3 and SYT7 to AR at cerebellar and hippocampal synapses. AR is dramatically reduced when both isoforms are absent, which alters the number and timing of postsynaptic action potentials. Our results confirm the long-standing prediction that SYT3 mediates AR and show that SYT3 and SYT7 act as dominant mechanisms for AR at three central synapses.
Collapse
Affiliation(s)
| | - Amita Shrestha
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Daniel J Orlin
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Chloé L Le Moing
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Luke A Borchardt
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Skyler L Jackman
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
8
|
Guiberson NGL, Black LS, Haller JE, Brukner A, Abramov D, Ahmad S, Xie YX, Sharma M, Burré J. Disease-linked mutations in Munc18-1 deplete synaptic Doc2. Brain 2024; 147:2185-2202. [PMID: 38242640 PMCID: PMC11146428 DOI: 10.1093/brain/awae019] [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: 05/31/2022] [Revised: 11/20/2023] [Accepted: 01/09/2024] [Indexed: 01/21/2024] Open
Abstract
Heterozygous de novo mutations in the neuronal protein Munc18-1/STXBP1 cause syndromic neurological symptoms, including severe epilepsy, intellectual disability, developmental delay, ataxia and tremor, summarized as STXBP1 encephalopathies. Although haploinsufficiency is the prevailing disease mechanism, it remains unclear how the reduction in Munc18-1 levels causes synaptic dysfunction in disease as well as how haploinsufficiency alone can account for the significant heterogeneity among patients in terms of the presence, onset and severity of different symptoms. Using biochemical and cell biological readouts on mouse brains, cultured mouse neurons and heterologous cells, we found that the synaptic Munc18-1 interactors Doc2A and Doc2B are unstable in the absence of Munc18-1 and aggregate in the presence of disease-causing Munc18-1 mutants. In haploinsufficiency-mimicking heterozygous knockout neurons, we found a reduction in Doc2A/B levels that is further aggravated by the presence of the disease-causing Munc18-1 mutation G544D as well as an impairment in Doc2A/B synaptic targeting in both genotypes. We also demonstrated that overexpression of Doc2A/B partially rescues synaptic dysfunction in heterozygous knockout neurons but not heterozygous knockout neurons expressing G544D Munc18-1. Our data demonstrate that STXBP1 encephalopathies are not only characterized by the dysfunction of Munc18-1 but also by the dysfunction of the Munc18-1 binding partners Doc2A and Doc2B, and that this dysfunction is exacerbated by the presence of a Munc18-1 missense mutant. These findings may offer a novel explanation for the significant heterogeneity in symptoms observed among STXBP1 encephalopathy patients.
Collapse
Affiliation(s)
- Noah Guy Lewis Guiberson
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Luca S Black
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jillian E Haller
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Aniv Brukner
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Debra Abramov
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Saad Ahmad
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Yan Xin Xie
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Manu Sharma
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jacqueline Burré
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
9
|
Zhang H, Lei M, Zhang Y, Li H, He Z, Xie S, Zhu L, Wang S, Liu J, Li Y, Lu Y, Ma C. Phosphorylation of Doc2 by EphB2 modulates Munc13-mediated SNARE complex assembly and neurotransmitter release. SCIENCE ADVANCES 2024; 10:eadi7024. [PMID: 38758791 PMCID: PMC11100570 DOI: 10.1126/sciadv.adi7024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 04/12/2024] [Indexed: 05/19/2024]
Abstract
At the synapse, presynaptic neurotransmitter release is tightly controlled by release machinery, involving the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins and Munc13. The Ca2+ sensor Doc2 cooperates with Munc13 to regulate neurotransmitter release, but the underlying mechanisms remain unclear. In our study, we have characterized the binding mode between Doc2 and Munc13 and found that Doc2 originally occludes Munc13 to inhibit SNARE complex assembly. Moreover, our investigation unveiled that EphB2, a presynaptic adhesion molecule (SAM) with inherent tyrosine kinase functionality, exhibits the capacity to phosphorylate Doc2. This phosphorylation attenuates Doc2 block on Munc13 to promote SNARE complex assembly, which functionally induces spontaneous release and synaptic augmentation. Consistently, application of a Doc2 peptide that interrupts Doc2-Munc13 interplay impairs excitatory synaptic transmission and leads to dysfunction in spatial learning and memory. These data provide evidence that SAMs modulate neurotransmitter release by controlling SNARE complex assembly.
Collapse
Affiliation(s)
- Hong Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China
| | - Mengshi Lei
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China
| | - Yu Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China
| | - Hao Li
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhen He
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Sheng Xie
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China
| | - Le Zhu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China
| | - Shen Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China
| | - Jianfeng Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China
| | - Yan Li
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Youming Lu
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Cong Ma
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
10
|
Bose D, Bera M, Norman CA, Timofeeva Y, Volynski KE, Krishnakumar SS. A minimal presynaptic protein machinery mediating synchronous and asynchronous exocytosis and short-term plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589559. [PMID: 38659918 PMCID: PMC11042279 DOI: 10.1101/2024.04.15.589559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Neurotransmitters are released from synaptic vesicles with remarkable precision in response to presynaptic Ca2+ influx but exhibit significant heterogeneity in exocytosis timing and efficacy based on the recent history of activity. This heterogeneity is critical for information transfer in the brain, yet its molecular basis remains poorly understood. Here, we employ a biochemically-defined fusion assay under physiologically-relevant conditions to delineate the minimal protein machinery sufficient to account for different modes of Ca2+-triggered vesicle fusion and short-term facilitation. We find that Synaptotagmin-1, Synaptotagmin-7, and Complexin, synergistically restrain SNARE complex assembly, thus preserving vesicles in a stably docked state at rest. Upon Ca2+ activation, Synaptotagmin-1 induces rapid vesicle fusion, while Synaptotagmin-7 mediates delayed fusion. Competitive binding of Synaptotagmin-1 and Synaptotagmin-7 to the same SNAREs, coupled with differential rates of Ca2+-triggered fusion clamp reversal, govern the kinetics of vesicular fusion. Under conditions mimicking sustained neuronal activity, the Synaptotagmin-7 fusion clamp is destabilized by the elevated basal Ca2+ concentration, thereby enhancing the synchronous component of fusion. These findings provide a direct demonstration that a small set of proteins is sufficient to account for how nerve terminals adapt and regulate the Ca2+-evoked neurotransmitter exocytosis process to support their specialized functions in the nervous system.
Collapse
Affiliation(s)
- Dipayan Bose
- Yale Nanobiology Institute, Yale University School of Medicine, New Haven, USA
- Department of Neurology, Yale University School of Medicine, New Haven, USA
| | - Manindra Bera
- Yale Nanobiology Institute, Yale University School of Medicine, New Haven, USA
- Cell Biology, Yale University School of Medicine, New Haven, USA
| | - Chris A Norman
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, UK
- Department of Computer Science, University of Warwick, Coventry, UK
| | - Yulia Timofeeva
- Department of Computer Science, University of Warwick, Coventry, UK
| | - Kirill E Volynski
- Cell Biology, Yale University School of Medicine, New Haven, USA
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, UK
| | - Shyam S Krishnakumar
- Yale Nanobiology Institute, Yale University School of Medicine, New Haven, USA
- Department of Neurology, Yale University School of Medicine, New Haven, USA
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, UK
| |
Collapse
|
11
|
Wu Z, Kusick GF, Berns MMM, Raychaudhuri S, Itoh K, Walter AM, Chapman ER, Watanabe S. Synaptotagmin 7 docks synaptic vesicles to support facilitation and Doc2α-triggered asynchronous release. eLife 2024; 12:RP90632. [PMID: 38536730 PMCID: PMC10972563 DOI: 10.7554/elife.90632] [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] [Indexed: 04/11/2024] Open
Abstract
Despite decades of intense study, the molecular basis of asynchronous neurotransmitter release remains enigmatic. Synaptotagmin (syt) 7 and Doc2 have both been proposed as Ca2+ sensors that trigger this mode of exocytosis, but conflicting findings have led to controversy. Here, we demonstrate that at excitatory mouse hippocampal synapses, Doc2α is the major Ca2+ sensor for asynchronous release, while syt7 supports this process through activity-dependent docking of synaptic vesicles. In synapses lacking Doc2α, asynchronous release after single action potentials is strongly reduced, while deleting syt7 has no effect. However, in the absence of syt7, docked vesicles cannot be replenished on millisecond timescales. Consequently, both synchronous and asynchronous release depress from the second pulse onward during repetitive activity. By contrast, synapses lacking Doc2α have normal activity-dependent docking, but continue to exhibit decreased asynchronous release after multiple stimuli. Moreover, disruption of both Ca2+ sensors is non-additive. These findings result in a new model whereby syt7 drives activity-dependent docking, thus providing synaptic vesicles for synchronous (syt1) and asynchronous (Doc2 and other unidentified sensors) release during ongoing transmission.
Collapse
Affiliation(s)
- Zhenyong Wu
- Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
- Howard Hughes Medical InstituteMadisonUnited States
| | - Grant F Kusick
- Department of Cell Biology, Johns Hopkins University, School of MedicineBaltimoreUnited States
- Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Manon MM Berns
- Department of Neuroscience, University of CopenhagenCopenhagenDenmark
| | - Sumana Raychaudhuri
- Department of Cell Biology, Johns Hopkins University, School of MedicineBaltimoreUnited States
| | - Kie Itoh
- Department of Cell Biology, Johns Hopkins University, School of MedicineBaltimoreUnited States
| | - Alexander M Walter
- Department of Neuroscience, University of CopenhagenCopenhagenDenmark
- Molecular and Theoretical Neuroscience, Leibniz-Institut für Molekulare Pharmakologie, FMP im CharitéCrossOverBerlinGermany
| | - Edwin R Chapman
- Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
- Howard Hughes Medical InstituteMadisonUnited States
| | - Shigeki Watanabe
- Department of Cell Biology, Johns Hopkins University, School of MedicineBaltimoreUnited States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
12
|
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: 0.5] [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
|
13
|
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
|
14
|
Tu Y, Qin J, Zhang QM, Tang TS, Wang L, Yao J. Secretagogin regulates asynchronous and spontaneous glutamate release in hippocampal neurons through interaction with Doc2α. LIFE MEDICINE 2023; 2:lnad041. [PMID: 39872889 PMCID: PMC11749858 DOI: 10.1093/lifemedi/lnad041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/13/2023] [Indexed: 01/30/2025]
Abstract
Synaptic vesicle (SV) exocytosis is orchestrated by protein machineries consisting of the SNARE complex, Ca2+ sensors, and their partners. Secretagogin (SCGN) is a Ca2+-binding protein involved in multiple forms of vesicle secretion. Although SCGN is implicated in multiple neurological disorders, its role in SV exocytosis in neurons remains unknown. Here, using knockout and knockdown techniques, we report that SCGN could regulate the asynchronous and spontaneous forms of excitatory but not inhibitory SV exocytosis in mouse hippocampal neurons. Furthermore, SCGN functioned in glutamate release via directly interacting with Doc2α, a high-affinity Ca2+ sensor specific for asynchronous and spontaneous SV exocytosis. Conversely, the interaction with SCGN is also required for Doc2α to execute its Ca2+ sensor function in SV release. Together, our study revealed that SCGN plays an important role in asynchronous and spontaneous glutamate release through its interaction with Doc2α.
Collapse
Affiliation(s)
- Yingfeng Tu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jiao Qin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Qiao-Ming Zhang
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Tie-Shan Tang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing Institute for Stem Cell and Regenerative Medicine, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lifang Wang
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing 100191, China
| | - Jun Yao
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
15
|
Jusyte M, Blaum N, Böhme MA, Berns MMM, Bonard AE, Vámosi ÁB, Pushpalatha KV, Kobbersmed JRL, Walter AM. Unc13A dynamically stabilizes vesicle priming at synaptic release sites for short-term facilitation and homeostatic potentiation. Cell Rep 2023; 42:112541. [PMID: 37243591 DOI: 10.1016/j.celrep.2023.112541] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/10/2023] [Accepted: 05/03/2023] [Indexed: 05/29/2023] Open
Abstract
Presynaptic plasticity adjusts neurotransmitter (NT) liberation. Short-term facilitation (STF) tunes synapses to millisecond repetitive activation, while presynaptic homeostatic potentiation (PHP) of NT release stabilizes transmission over minutes. Despite different timescales of STF and PHP, our analysis of Drosophila neuromuscular junctions reveals functional overlap and shared molecular dependence on the release-site protein Unc13A. Mutating Unc13A's calmodulin binding domain (CaM-domain) increases baseline transmission while blocking STF and PHP. Mathematical modeling suggests that Ca2+/calmodulin/Unc13A interaction plastically stabilizes vesicle priming at release sites and that CaM-domain mutation causes constitutive stabilization, thereby blocking plasticity. Labeling the functionally essential Unc13A MUN domain reveals higher STED microscopy signals closer to release sites following CaM-domain mutation. Acute phorbol ester treatment similarly enhances NT release and blocks STF/PHP in synapses expressing wild-type Unc13A, while CaM-domain mutation occludes this, indicating common downstream effects. Thus, Unc13A regulatory domains integrate signals across timescales to switch release-site participation for synaptic plasticity.
Collapse
Affiliation(s)
- Meida Jusyte
- Molecular and Theoretical Neuroscience, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany; Einstein Center for Neurosciences Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Natalie Blaum
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Mathias A Böhme
- Molecular and Theoretical Neuroscience, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany; Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Manon M M Berns
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Alix E Bonard
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Ábel B Vámosi
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | | | - Janus R L Kobbersmed
- Department of Mathematical Sciences, University of Copenhagen, Copenhagen, Denmark; Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Alexander M Walter
- Molecular and Theoretical Neuroscience, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany; Einstein Center for Neurosciences Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany; Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
16
|
Wang QW, Qin J, Chen YF, Tu Y, Xing YY, Wang Y, Yang LY, Lu SY, Geng L, Shi W, Yang Y, Yao J. 16p11.2 CNV gene Doc2α functions in neurodevelopment and social behaviors through interaction with Secretagogin. Cell Rep 2023; 42:112691. [PMID: 37354460 DOI: 10.1016/j.celrep.2023.112691] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 04/22/2023] [Accepted: 06/08/2023] [Indexed: 06/26/2023] Open
Abstract
Copy-number variations (CNVs) of the human 16p11.2 genetic locus are associated with neurodevelopmental disorders, including autism spectrum disorders (ASDs) and schizophrenia. However, it remains largely unclear how this locus is involved in the disease pathogenesis. Doc2α is localized within this locus. Here, using in vivo and ex vivo electrophysiological and morphological approaches, we show that Doc2α-deficient mice have neuronal morphological abnormalities and defects in neural activity. Moreover, the Doc2α-deficient mice exhibit social and repetitive behavioral deficits. Furthermore, we demonstrate that Doc2α functions in behavioral and neural phenotypes through interaction with Secretagogin (SCGN). Finally, we demonstrate that SCGN functions in social/repetitive behaviors, glutamate release, and neuronal morphology of the mice through its Doc2α-interacting activity. Therefore, Doc2α likely contributes to neurodevelopmental disorders through its interaction with SCGN.
Collapse
Affiliation(s)
- Qiu-Wen Wang
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Junhong Qin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China; School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yan-Fen Chen
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yingfeng Tu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China; School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yun-Yun Xing
- Jiangsu Key Laboratory of Language and Cognitive Neuroscience, School of Linguistic Sciences and Arts, Jiangsu Normal University, Xuzhou 221116, China; Jiangsu Collaborative Innovation Center for Language Ability, Xuzhou 221009, China
| | - Yuchen Wang
- School of Engineering Medicine and School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Lv-Yu Yang
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Si-Yao Lu
- Jiangsu Key Laboratory of Language and Cognitive Neuroscience, School of Linguistic Sciences and Arts, Jiangsu Normal University, Xuzhou 221116, China; Jiangsu Collaborative Innovation Center for Language Ability, Xuzhou 221009, China
| | - Libo Geng
- Jiangsu Key Laboratory of Language and Cognitive Neuroscience, School of Linguistic Sciences and Arts, Jiangsu Normal University, Xuzhou 221116, China; Jiangsu Collaborative Innovation Center for Language Ability, Xuzhou 221009, China
| | - Wei Shi
- School of Engineering Medicine and School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.
| | - Yiming Yang
- Jiangsu Key Laboratory of Language and Cognitive Neuroscience, School of Linguistic Sciences and Arts, Jiangsu Normal University, Xuzhou 221116, China; Jiangsu Collaborative Innovation Center for Language Ability, Xuzhou 221009, China.
| | - Jun Yao
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
17
|
Ogunmowo TH, Jing H, Raychaudhuri S, Kusick GF, Imoto Y, Li S, Itoh K, Ma Y, Jafri H, Dalva MB, Chapman ER, Ha T, Watanabe S, Liu J. Membrane compression by synaptic vesicle exocytosis triggers ultrafast endocytosis. Nat Commun 2023; 14:2888. [PMID: 37210439 PMCID: PMC10199930 DOI: 10.1038/s41467-023-38595-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 05/09/2023] [Indexed: 05/22/2023] Open
Abstract
Compensatory endocytosis keeps the membrane surface area of secretory cells constant following exocytosis. At chemical synapses, clathrin-independent ultrafast endocytosis maintains such homeostasis. This endocytic pathway is temporally and spatially coupled to exocytosis; it initiates within 50 ms at the region immediately next to the active zone where vesicles fuse. However, the coupling mechanism is unknown. Here, we demonstrate that filamentous actin is organized as a ring, surrounding the active zone at mouse hippocampal synapses. Assuming the membrane area conservation is due to this actin ring, our theoretical model suggests that flattening of fused vesicles exerts lateral compression in the plasma membrane, resulting in rapid formation of endocytic pits at the border between the active zone and the surrounding actin-enriched region. Consistent with model predictions, our data show that ultrafast endocytosis requires sufficient compression by exocytosis of multiple vesicles and does not initiate when actin organization is disrupted, either pharmacologically or by ablation of the actin-binding protein Epsin1. Our work suggests that membrane mechanics underlie the rapid coupling of exocytosis to endocytosis at synapses.
Collapse
Affiliation(s)
- Tyler H Ogunmowo
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, US
- Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, US
- Biochemistry, Cellular and Molecular Biology graduate program, Johns Hopkins University, Baltimore, MD, US
| | - Haoyuan Jing
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, US
- Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, US
| | - Sumana Raychaudhuri
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, US
- Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, US
| | - Grant F Kusick
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, US
- Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, US
- Biochemistry, Cellular and Molecular Biology graduate program, Johns Hopkins University, Baltimore, MD, US
| | - Yuuta Imoto
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, US
- Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, US
| | - Shuo Li
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, US
- Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, US
- Department of Ophthalmology, School of Medicine, Stanford University, Palo Alto, CA, US
| | - Kie Itoh
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, US
- Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, US
| | - Ye Ma
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, US
| | - Haani Jafri
- Department of Neuroscience and Jefferson Synaptic Biology Center, Thomas Jefferson University, Philadelphia, PA, US
| | - Matthew B Dalva
- Department of Neuroscience and Jefferson Synaptic Biology Center, Thomas Jefferson University, Philadelphia, PA, US
- Department of Cell and Molecular Biology and the Tulane Brain Institute, Tulane University, New Orleans, LA, US
| | - Edwin R Chapman
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, US
- Howard Hughes Medical Institute, Madison, WI, US
| | - Taekjip Ha
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, US
- Department of Biophysics and Biophysical Chemistry, School of Medicine, Johns Hopkins University, Baltimore, MD, US
- Department of Biophysics, Johns Hopkins University, Baltimore, MD, US
- Howard Hughes Medical Institute, Baltimore, MD, US
| | - Shigeki Watanabe
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, US.
- Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, US.
- Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, US.
| | - Jian Liu
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, MD, US.
- Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, US.
| |
Collapse
|
18
|
Lebowitz JJ, Banerjee A, Qiao C, Bunzow JR, Williams JT, Kaeser PS. Synaptotagmin-1 is a Ca 2+ sensor for somatodendritic dopamine release. Cell Rep 2023; 42:111915. [PMID: 36640316 PMCID: PMC9993464 DOI: 10.1016/j.celrep.2022.111915] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 11/07/2022] [Accepted: 12/13/2022] [Indexed: 12/31/2022] Open
Abstract
Modes of somatodendritic transmission range from rapid synaptic signaling to protracted regulation over distance. Somatodendritic dopamine secretion in the midbrain leads to D2 receptor-induced modulation of dopamine neurons on the timescale of seconds. Temporally imprecise release mechanisms are often presumed to be at play, and previous work indeed suggested roles for slow Ca2+ sensors. We here use mouse genetics and whole-cell electrophysiology to establish that the fast Ca2+ sensor synaptotagmin-1 (Syt-1) is important for somatodendritic dopamine release. Syt-1 ablation from dopamine neurons strongly reduces stimulus-evoked D2 receptor-mediated inhibitory postsynaptic currents (D2-IPSCs) in the midbrain. D2-IPSCs evoked by paired stimuli exhibit less depression, and high-frequency trains restore dopamine release. Spontaneous somatodendritic dopamine secretion is independent of Syt-1, supporting that its exocytotic mechanisms differ from evoked release. We conclude that somatodendritic dopamine transmission relies on the fast Ca2+ sensor Syt-1, leading to synchronous release in response to the initial stimulus.
Collapse
Affiliation(s)
- Joseph J Lebowitz
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Aditi Banerjee
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Claire Qiao
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - James R Bunzow
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - John T Williams
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA.
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
19
|
Yang X, Tu W, Gao X, Zhang Q, Guan J, Zhang J. Functional regulation of syntaxin-1: An underlying mechanism mediating exocytosis in neuroendocrine cells. Front Endocrinol (Lausanne) 2023; 14:1096365. [PMID: 36742381 PMCID: PMC9892835 DOI: 10.3389/fendo.2023.1096365] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 01/05/2023] [Indexed: 01/20/2023] Open
Abstract
The fusion of the secretory vesicle with the plasma membrane requires the assembly of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein complexes formed by synaptobrevin, syntaxin-1, and SNAP-25. Within the pathway leading to exocytosis, the transitions between the "open" and "closed" conformations of syntaxin-1 function as a switch for the fusion of vesicles with the plasma membranes; rapid assembly and disassembly of syntaxin-1 clusters on the plasma membrane provide docking and fusion sites for secretory vesicles in neuroendocrine cells; and the fully zippered trans-SNARE complex, which requires the orderly, rapid and accurate binding of syntaxin-1 to other SNARE proteins, play key roles in triggering fusion. All of these reactions that affect exocytosis under physiological conditions are tightly regulated by multiple factors. Here, we review the current evidence for the involvement of syntaxin-1 in the mechanism of neuroendocrine cell exocytosis, discuss the roles of multiple factors such as proteins, lipids, protein kinases, drugs, and toxins in SNARE complex-mediated membrane fusion, and present an overview of syntaxin-1 mutation-associated diseases with a view to developing novel mechanistic therapeutic targets for the treatment of neuroendocrine disorders.
Collapse
Affiliation(s)
- Xinquan Yang
- Anesthesia and Perioperative Medicine laboratory, the Affiliated Lianyungang Hospital of Jiangsu University, Lianyungang, China
| | - Weifeng Tu
- Faculty of Anesthesioloy, Suzhou Hospital Affiliated to Medical School of Nanjing University, Suzhou, China
| | - Xuzhu Gao
- Department of Central Laboratory, Lianyungang Hospital Affiliated to Jiangsu University, Lianyungang, China
| | - Qi Zhang
- Anesthesia and Perioperative Medicine laboratory, the Affiliated Lianyungang Hospital of Jiangsu University, Lianyungang, China
| | - Jinping Guan
- Anesthesia and Perioperative Medicine laboratory, the Affiliated Lianyungang Hospital of Jiangsu University, Lianyungang, China
| | - Junlong Zhang
- Anesthesia and Perioperative Medicine laboratory, the Affiliated Lianyungang Hospital of Jiangsu University, Lianyungang, China
- *Correspondence: Junlong Zhang,
| |
Collapse
|
20
|
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
|
21
|
Cui L, Li H, Xi Y, Hu Q, Liu H, Fan J, Xiang Y, Zhang X, Shui W, Lai Y. Vesicle trafficking and vesicle fusion: mechanisms, biological functions, and their implications for potential disease therapy. MOLECULAR BIOMEDICINE 2022; 3:29. [PMID: 36129576 PMCID: PMC9492833 DOI: 10.1186/s43556-022-00090-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/12/2022] [Indexed: 11/10/2022] Open
Abstract
Intracellular vesicle trafficking is the fundamental process to maintain the homeostasis of membrane-enclosed organelles in eukaryotic cells. These organelles transport cargo from the donor membrane to the target membrane through the cargo containing vesicles. Vesicle trafficking pathway includes vesicle formation from the donor membrane, vesicle transport, and vesicle fusion with the target membrane. Coat protein mediated vesicle formation is a delicate membrane budding process for cargo molecules selection and package into vesicle carriers. Vesicle transport is a dynamic and specific process for the cargo containing vesicles translocation from the donor membrane to the target membrane. This process requires a group of conserved proteins such as Rab GTPases, motor adaptors, and motor proteins to ensure vesicle transport along cytoskeletal track. Soluble N-ethyl-maleimide-sensitive factor (NSF) attachment protein receptors (SNARE)-mediated vesicle fusion is the final process for vesicle unloading the cargo molecules at the target membrane. To ensure vesicle fusion occurring at a defined position and time pattern in eukaryotic cell, multiple fusogenic proteins, such as synaptotagmin (Syt), complexin (Cpx), Munc13, Munc18 and other tethering factors, cooperate together to precisely regulate the process of vesicle fusion. Dysfunctions of the fusogenic proteins in SNARE-mediated vesicle fusion are closely related to many diseases. Recent studies have suggested that stimulated membrane fusion can be manipulated pharmacologically via disruption the interface between the SNARE complex and Ca2+ sensor protein. Here, we summarize recent insights into the molecular mechanisms of vesicle trafficking, and implications for the development of new therapeutics based on the manipulation of vesicle fusion.
Collapse
|
22
|
Xiao J, Meng X, Chen K, Wang J, Wu L, Chen Y, Yu X, Feng J, Li Z. Down-Regulation of Double C2 Domain Alpha Promotes the Formation of Hyperplastic Nerve Fibers in Aganglionic Segments of Hirschsprung's Disease. Int J Mol Sci 2022; 23:10204. [PMID: 36142117 PMCID: PMC9499397 DOI: 10.3390/ijms231810204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/22/2022] Open
Abstract
Hirschsprung's disease (HSCR) is a common developmental anomaly of the gastrointestinal tract in children. The most significant characteristics of aganglionic segments in HSCR are hyperplastic extrinsic nerve fibers and the absence of endogenous ganglion plexus. Double C2 domain alpha (DOC2A) is mainly located in the nucleus and is involved in Ca2+-dependent neurotransmitter release. The loss function of DOC2A influences postsynaptic protein synthesis, dendrite morphology, postsynaptic receptor density and synaptic plasticity. It is still unknown why hyperplastic extrinsic nerve fibers grow into aganglionic segments in HSCR. We detected the expression of DOC2A in HSCR aganglionic segment colons and established three DOC2A-knockdown models in the Neuro-2a cell line, neural spheres and zebrafish separately. First, we detected the protein and mRNA expression of DOC2A and found that DOC2A was negatively correlated with AChE+ grades. Second, in the Neuro-2a cell lines, we found that the amount of neurite outgrowth and mean area per cell were significantly increased, which suggested that the inhibition of DOC2A promotes nerve fiber formation and the neuron's polarity. In the neural spheres, we found that the DOC2A knockdown was manifested by a more obvious connection of nerve fibers in neural spheres. Then, we knocked down Doc2a in zebrafish and found that the down-regulation of Doc2a accelerates the formation of hyperplastic nerve fibers in aganglionic segments in zebrafish. Finally, we detected the expression of MUNC13-2 (UNC13B), which was obviously up-regulated in Grade3/4 (lower DOC2A expression) compared with Grade1/2 (higher DOC2A expression) in the circular muscle layer and longitudinal muscle layer. The expression of UNC13B was up-regulated with the knocking down of DOC2A, and there were protein interactions between DOC2A and UNC13B. The down-regulation of DOC2A may be an important factor leading to hyperplastic nerve fibers in aganglionic segments of HSCR. UNC13B seems to be a downstream molecule to DOC2A, which may participate in the spasm of aganglionic segments of HSCR patient colons.
Collapse
Affiliation(s)
- Jun Xiao
- Department of Pediatric Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Clinical Center of Hirschsprung’s Disease and Allied Disorders, Wuhan 430030, China
| | - Xinyao Meng
- Department of Pediatric Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Clinical Center of Hirschsprung’s Disease and Allied Disorders, Wuhan 430030, China
| | - Ke Chen
- Department of Pediatric Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Clinical Center of Hirschsprung’s Disease and Allied Disorders, Wuhan 430030, China
| | - Jing Wang
- Department of Pediatric Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Clinical Center of Hirschsprung’s Disease and Allied Disorders, Wuhan 430030, China
| | - Luyao Wu
- Department of Pediatric Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Clinical Center of Hirschsprung’s Disease and Allied Disorders, Wuhan 430030, China
| | - Yingjian Chen
- Department of Pediatric Surgery, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Xiaosi Yu
- Department of Pediatric Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Clinical Center of Hirschsprung’s Disease and Allied Disorders, Wuhan 430030, China
| | - Jiexiong Feng
- Department of Pediatric Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Clinical Center of Hirschsprung’s Disease and Allied Disorders, Wuhan 430030, China
| | - Zhi Li
- Department of Pediatric Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Clinical Center of Hirschsprung’s Disease and Allied Disorders, Wuhan 430030, China
| |
Collapse
|
23
|
Kobbersmed JRL, Berns MMM, Ditlevsen S, Sørensen JB, Walter AM. Allosteric stabilization of calcium and phosphoinositide dual binding engages several synaptotagmins in fast exocytosis. eLife 2022; 11:74810. [PMID: 35929728 PMCID: PMC9489213 DOI: 10.7554/elife.74810] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 08/04/2022] [Indexed: 12/04/2022] Open
Abstract
Synaptic communication relies on the fusion of synaptic vesicles with the plasma membrane, which leads to neurotransmitter release. This exocytosis is triggered by brief and local elevations of intracellular Ca2+ with remarkably high sensitivity. How this is molecularly achieved is unknown. While synaptotagmins confer the Ca2+ sensitivity of neurotransmitter exocytosis, biochemical measurements reported Ca2+ affinities too low to account for synaptic function. However, synaptotagmin’s Ca2+ affinity increases upon binding the plasma membrane phospholipid PI(4,5)P2 and, vice versa, Ca2+ binding increases synaptotagmin’s PI(4,5)P2 affinity, indicating a stabilization of the Ca2+/PI(4,5)P2 dual-bound state. Here, we devise a molecular exocytosis model based on this positive allosteric stabilization and the assumptions that (1.) synaptotagmin Ca2+/PI(4,5)P2 dual binding lowers the energy barrier for vesicle fusion and that (2.) the effect of multiple synaptotagmins on the energy barrier is additive. The model, which relies on biochemically measured Ca2+/PI(4,5)P2 affinities and protein copy numbers, reproduced the steep Ca2+ dependency of neurotransmitter release. Our results indicate that each synaptotagmin engaging in Ca2+/PI(4,5)P2 dual-binding lowers the energy barrier for vesicle fusion by ~5 kBT and that allosteric stabilization of this state enables the synchronized engagement of several (typically three) synaptotagmins for fast exocytosis. Furthermore, we show that mutations altering synaptotagmin’s allosteric properties may show dominant-negative effects, even though synaptotagmins act independently on the energy barrier, and that dynamic changes of local PI(4,5)P2 (e.g. upon vesicle movement) dramatically impact synaptic responses. We conclude that allosterically stabilized Ca2+/PI(4,5)P2 dual binding enables synaptotagmins to exert their coordinated function in neurotransmission. For our brains and nervous systems to work properly, the nerve cells within them must be able to ‘talk’ to each other. They do this by releasing chemical signals called neurotransmitters which other cells can detect and respond to. Neurotransmitters are packaged in tiny membrane-bound spheres called vesicles. When a cell of the nervous system needs to send a signal to its neighbours, the vesicles fuse with the outer membrane of the cell, discharging their chemical contents for other cells to detect. The initial trigger for neurotransmitter release is a short, fast increase in the amount of calcium ions inside the signalling cell. One of the main proteins that helps regulate this process is synaptotagmin which binds to calcium and gives vesicles the signal to start unloading their chemicals. Despite acting as a calcium sensor, synaptotagmin actually has a very low affinity for calcium ions by itself, meaning that it would not be efficient for the protein to respond alone. Synpatotagmin is more likely to bind to calcium if it is attached to a molecule called PIP2, which is found in the membranes of cells The effect also occurs in reverse, as the binding of calcium to synaptotagmin increases the protein’s affinity for PIP2. However, how these three molecules – synaptotagmin, PIP2, and calcium – work together to achieve the physiological release of neurotransmitters is poorly understood. To help answer this question, Kobbersmed, Berns et al. set up a computer simulation of ‘virtual vesicles’ using available experimental data on synaptotagmin’s affinity with calcium and PIP2. In this simulation, synaptotagmin could only trigger the release of neurotransmitters when bound to both calcium and PIP2. The model also showed that each ‘complex’ of synaptotagmin/calcium/PIP2 made the vesicles more likely to fuse with the outer membrane of the cell – to the extent that only a handful of synaptotagmin molecules were needed to start neurotransmitter release from a single vesicle. These results shed new light on a biological process central to the way nerve cells communicate with each other. In the future, Kobbersmed, Berns et al. hope that this insight will help us to understand the cause of diseases where communication in the nervous system is impaired.
Collapse
Affiliation(s)
- Janus R L Kobbersmed
- Department of Mathematical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Manon M M Berns
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Ditlevsen
- Department of Mathematical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Alexander M Walter
- Department of Mathematical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
24
|
Mochida S. Mechanisms of Synaptic Vesicle Exo- and Endocytosis. Biomedicines 2022; 10:1593. [PMID: 35884898 PMCID: PMC9313035 DOI: 10.3390/biomedicines10071593] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 01/05/2023] Open
Abstract
Within 1 millisecond of action potential arrival at presynaptic terminals voltage-gated Ca2+ channels open. The Ca2+ channels are linked to synaptic vesicles which are tethered by active zone proteins. Ca2+ entrance into the active zone triggers: (1) the fusion of the vesicle and exocytosis, (2) the replenishment of the active zone with vesicles for incoming exocytosis, and (3) various types of endocytosis for vesicle reuse, dependent on the pattern of firing. These time-dependent vesicle dynamics are controlled by presynaptic Ca2+ sensor proteins, regulating active zone scaffold proteins, fusion machinery proteins, motor proteins, endocytic proteins, several enzymes, and even Ca2+ channels, following the decay of Ca2+ concentration after the action potential. Here, I summarize the Ca2+-dependent protein controls of synchronous and asynchronous vesicle release, rapid replenishment of the active zone, endocytosis, and short-term plasticity within 100 msec after the action potential. Furthermore, I discuss the contribution of active zone proteins to presynaptic plasticity and to homeostatic readjustment during and after intense activity, in addition to activity-dependent endocytosis.
Collapse
Affiliation(s)
- Sumiko Mochida
- Department of Physiology, Tokyo Medical University, Tokyo 160-8402, Japan
| |
Collapse
|
25
|
Egashira Y, Kumade A, Ojida A, Ono F. Spontaneously Recycling Synaptic Vesicles Constitute Readily Releasable Vesicles in Intact Neuromuscular Synapses. J Neurosci 2022; 42:3523-3536. [PMID: 35332083 PMCID: PMC9053852 DOI: 10.1523/jneurosci.2005-21.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 12/14/2022] Open
Abstract
Emerging evidence shows that spontaneous synaptic transmission plays crucial roles on neuronal functions through presynaptic molecular mechanisms distinct from that of action potential (AP)-evoked transmission. However, whether the synaptic vesicle (SV) population undergoing the two forms of transmission is segregated remains controversial due in part to the conflicting results observed in cultured neurons. Here we address this issue in intact neuromuscular synapses using transgenic zebrafish larvae expressing two different indicators targeted in the SVs: a pH-sensitive fluorescent protein, pHluorin, and a tag protein, HaloTag. By establishing a quantitative measure of recycled SV fractions, we found that ∼85% of SVs were mobilized by high-frequency AP firings. In contrast, spontaneously recycling SVs were mobilized only from <8% of SVs with a time constant of 45 min at 25°C, although prolonged AP inhibition mobilized an additional population with a delayed onset. The mobilization of the early-onset population was less temperature-sensitive and resistant to tetanus toxin, whereas that of the late-onset population was more sensitive to temperature and was inhibited by tetanus toxin, indicating that prolonged AP inhibition activated a distinct molecular machinery for spontaneous SV fusion. Therefore, the early-onset population limited to <8% was likely the only source of spontaneous release that occurred physiologically. We further showed that this limited population was independent from those reluctant to fuse during AP firing and was used in both the hypertonic stimulation and the immediate phase of AP-evoked releases, thereby matching the characteristics of the readily releasable pool.SIGNIFICANCE STATEMENT Synaptic vesicles (SVs) are divided into functionally distinct pools depending on how they respond to action potential (AP) firing. The origin of SVs used for spontaneous fusion remains enigmatic despite intensive studies in cultured preparations. We addressed this question in intact neuromuscular synapses and provided two findings. First, prolonged AP inhibition activated a distinct population of fusion, which needs to be distinguished from genuine spontaneous fusion arising from a highly limited fraction. Second, the limited fraction observed early in the AP inhibition period exhibited the characteristics of readily releasable pool in the subsequent round of stimulation. Our study revealed that the origin of spontaneous SV fusion is restricted to the readily releasable pool among the SV pools involved in AP-evoked fusion.
Collapse
Affiliation(s)
- Yoshihiro Egashira
- Department of Physiology, Osaka Medical and Pharmaceutical University, Takatsuki, 569-8686, Japan
| | - Ayane Kumade
- Graduate School of Pharmaceutical Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Akio Ojida
- Graduate School of Pharmaceutical Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Fumihito Ono
- Department of Physiology, Osaka Medical and Pharmaceutical University, Takatsuki, 569-8686, Japan
| |
Collapse
|
26
|
Bisio H, Krishnan A, Marq JB, Soldati-Favre D. Toxoplasma gondii phosphatidylserine flippase complex ATP2B-CDC50.4 critically participates in microneme exocytosis. PLoS Pathog 2022; 18:e1010438. [PMID: 35325010 PMCID: PMC8982854 DOI: 10.1371/journal.ppat.1010438] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 04/05/2022] [Accepted: 03/11/2022] [Indexed: 12/22/2022] Open
Abstract
Regulated microneme secretion governs motility, host cell invasion and egress in the obligate intracellular apicomplexans. Intracellular calcium oscillations and phospholipid dynamics critically regulate microneme exocytosis. Despite its importance for the lytic cycle of these parasites, molecular mechanistic details about exocytosis are still missing. Some members of the P4-ATPases act as flippases, changing the phospholipid distribution by translocation from the outer to the inner leaflet of the membrane. Here, the localization and function of the repertoire of P4-ATPases was investigated across the lytic cycle of Toxoplasma gondii. Of relevance, ATP2B and the non-catalytic subunit cell division control protein 50.4 (CDC50.4) form a stable heterocomplex at the parasite plasma membrane, essential for microneme exocytosis. This complex is responsible for flipping phosphatidylserine, which presumably acts as a lipid mediator for organelle fusion with the plasma membrane. Overall, this study points toward the importance of phosphatidylserine asymmetric distribution at the plasma membrane for microneme exocytosis. Biological membranes display diverse functions, including membrane fusion, which are conferred by a defined composition and organization of proteins and lipids. Apicomplexan parasites possess specialized secretory organelles (micronemes), implicated in motility, invasion and egress from host cells. Microneme exocytosis is already known to depend on phosphatidic acid for its fusion with the plasma membrane. Here we identify a type P4-ATPase and its CDC50 chaperone (ATP2B-CDC50.4) that act as a flippase and contribute to the enrichment of phosphatidylserine (PS) in the inner leaflet of the parasite plasma membrane. The disruption of PS asymmetric distribution at the plasma membrane impacts microneme exocytosis. Overall, our results shed light on the importance of membrane homeostasis and lipid composition in controlling microneme secretion.
Collapse
Affiliation(s)
- Hugo Bisio
- Department of Microbiology and Molecular Medicine, CMU, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Aarti Krishnan
- Department of Microbiology and Molecular Medicine, CMU, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jean-Baptiste Marq
- Department of Microbiology and Molecular Medicine, CMU, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, CMU, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- * E-mail:
| |
Collapse
|
27
|
Li L, Liu H, Krout M, Richmond JE, Wang Y, Bai J, Weeratunga S, Collins BM, Ventimiglia D, Yu Y, Xia J, Tang J, Liu J, Hu Z. A novel dual Ca2+ sensor system regulates Ca2+-dependent neurotransmitter release. J Cell Biol 2021; 220:211787. [PMID: 33570571 PMCID: PMC7883739 DOI: 10.1083/jcb.202008121] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 12/19/2020] [Accepted: 01/15/2021] [Indexed: 02/08/2023] Open
Abstract
Ca2+-dependent neurotransmitter release requires synaptotagmins as Ca2+ sensors to trigger synaptic vesicle (SV) exocytosis via binding of their tandem C2 domains—C2A and C2B—to Ca2+. We have previously demonstrated that SNT-1, a mouse synaptotagmin-1 (Syt1) homologue, functions as the fast Ca2+ sensor in Caenorhabditis elegans. Here, we report a new Ca2+ sensor, SNT-3, which triggers delayed Ca2+-dependent neurotransmitter release. snt-1;snt-3 double mutants abolish evoked synaptic transmission, demonstrating that C. elegans NMJs use a dual Ca2+ sensor system. SNT-3 possesses canonical aspartate residues in both C2 domains, but lacks an N-terminal transmembrane (TM) domain. Biochemical evidence demonstrates that SNT-3 binds both Ca2+ and the plasma membrane. Functional analysis shows that SNT-3 is activated when SNT-1 function is impaired, triggering SV release that is loosely coupled to Ca2+ entry. Compared with SNT-1, which is tethered to SVs, SNT-3 is not associated with SV. Eliminating the SV tethering of SNT-1 by removing the TM domain or the whole N terminus rescues fast release kinetics, demonstrating that cytoplasmic SNT-1 is still functional and triggers fast neurotransmitter release, but also exhibits decreased evoked amplitude and release probability. These results suggest that the fast and slow properties of SV release are determined by the intrinsically different C2 domains in SNT-1 and SNT-3, rather than their N-termini–mediated membrane tethering. Our findings therefore reveal a novel dual Ca2+ sensor system in C. elegans and provide significant insights into Ca2+-regulated exocytosis.
Collapse
Affiliation(s)
- Lei Li
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Haowen Liu
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Mia Krout
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL
| | - Janet E Richmond
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL
| | - Yu Wang
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Jihong Bai
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Saroja Weeratunga
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Brett M Collins
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Donovan Ventimiglia
- Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, New York, NY
| | - Yi Yu
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Jingyao Xia
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Jing Tang
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Jie Liu
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Zhitao Hu
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
28
|
Vevea JD, Kusick GF, Courtney KC, Chen E, Watanabe S, Chapman ER. Synaptotagmin 7 is targeted to the axonal plasma membrane through γ-secretase processing to promote synaptic vesicle docking in mouse hippocampal neurons. eLife 2021; 10:e67261. [PMID: 34543184 PMCID: PMC8452306 DOI: 10.7554/elife.67261] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 08/27/2021] [Indexed: 12/28/2022] Open
Abstract
Synaptotagmin 7 (SYT7) has emerged as a key regulator of presynaptic function, but its localization and precise role in the synaptic vesicle cycle remain the subject of debate. Here, we used iGluSnFR to optically interrogate glutamate release, at the single-bouton level, in SYT7KO-dissociated mouse hippocampal neurons. We analyzed asynchronous release, paired-pulse facilitation, and synaptic vesicle replenishment and found that SYT7 contributes to each of these processes to different degrees. 'Zap-and-freeze' electron microscopy revealed that a loss of SYT7 diminishes docking of synaptic vesicles after a stimulus and inhibits the recovery of depleted synaptic vesicles after a stimulus train. SYT7 supports these functions from the axonal plasma membrane, where its localization and stability require both γ-secretase-mediated cleavage and palmitoylation. In summary, SYT7 is a peripheral membrane protein that controls multiple modes of synaptic vesicle (SV) exocytosis and plasticity, in part, through enhancing activity-dependent docking of SVs.
Collapse
Affiliation(s)
- Jason D Vevea
- Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
- Howard Hughes Medical InstituteMadisonUnited States
| | - Grant F Kusick
- Department of Cell Biology, Johns Hopkins University, School of MedicineBaltimoreUnited States
- Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University, School of MedicineBaltimoreUnited States
| | - Kevin C Courtney
- Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
- Howard Hughes Medical InstituteMadisonUnited States
| | - Erin Chen
- Department of Cell Biology, Johns Hopkins University, School of MedicineBaltimoreUnited States
| | - Shigeki Watanabe
- Department of Cell Biology, Johns Hopkins University, School of MedicineBaltimoreUnited States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, School of MedicineBaltimoreUnited States
| | - Edwin R Chapman
- Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
- Howard Hughes Medical InstituteMadisonUnited States
| |
Collapse
|
29
|
Hays CL, Sladek AL, Thoreson WB. Resting and stimulated mouse rod photoreceptors show distinct patterns of vesicle release at ribbon synapses. J Gen Physiol 2021; 152:211528. [PMID: 33175961 PMCID: PMC7664508 DOI: 10.1085/jgp.202012716] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/01/2020] [Accepted: 10/19/2020] [Indexed: 01/04/2023] Open
Abstract
The vertebrate visual system can detect and transmit signals from single photons. To understand how single-photon responses are transmitted, we characterized voltage-dependent properties of glutamate release in mouse rods. We measured presynaptic glutamate transporter anion current and found that rates of synaptic vesicle release increased with voltage-dependent Ca2+ current. Ca2+ influx and release rate also rose with temperature, attaining a rate of ∼11 vesicles/s/ribbon at -40 mV (35°C). By contrast, spontaneous release events at hyperpolarized potentials (-60 to -70 mV) were univesicular and occurred at random intervals. However, when rods were voltage clamped at -40 mV for many seconds to simulate maintained darkness, release occurred in coordinated bursts of 17 ± 7 quanta (mean ± SD; n = 22). Like fast release evoked by brief depolarizing stimuli, these bursts involved vesicles in the readily releasable pool of vesicles and were triggered by the opening of nearby ribbon-associated Ca2+ channels. Spontaneous release rates were elevated and bursts were absent after genetic elimination of the Ca2+ sensor synaptotagmin 1 (Syt1). This study shows that at the resting potential in darkness, rods release glutamate-filled vesicles from a pool at the base of synaptic ribbons at low rates but in Syt1-dependent bursts. The absence of bursting in cones suggests that this behavior may have a role in transmitting scotopic responses.
Collapse
Affiliation(s)
- Cassandra L Hays
- Cellular and Integrative Physiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE.,Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Asia L Sladek
- Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Wallace B Thoreson
- Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE.,Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
30
|
Xue R, Meng H, Yin J, Xia J, Hu Z, Liu H. The Role of Calmodulin vs. Synaptotagmin in Exocytosis. Front Mol Neurosci 2021; 14:691363. [PMID: 34421537 PMCID: PMC8375295 DOI: 10.3389/fnmol.2021.691363] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/19/2021] [Indexed: 12/04/2022] Open
Abstract
Exocytosis is a Ca2+-regulated process that requires the participation of Ca2+ sensors. In the 1980s, two classes of Ca2+-binding proteins were proposed as putative Ca2+ sensors: EF-hand protein calmodulin, and the C2 domain protein synaptotagmin. In the next few decades, numerous studies determined that in the final stage of membrane fusion triggered by a micromolar boost in the level of Ca2+, the low affinity Ca2+-binding protein synaptotagmin, especially synaptotagmin 1 and 2, acts as the primary Ca2+ sensor, whereas calmodulin is unlikely to be functional due to its high Ca2+ affinity. However, in the meantime emerging evidence has revealed that calmodulin is involved in the earlier exocytotic steps prior to fusion, such as vesicle trafficking, docking and priming by acting as a high affinity Ca2+ sensor activated at submicromolar level of Ca2+. Calmodulin directly interacts with multiple regulatory proteins involved in the regulation of exocytosis, including VAMP, myosin V, Munc13, synapsin, GAP43 and Rab3, and switches on key kinases, such as type II Ca2+/calmodulin-dependent protein kinase, to phosphorylate a series of exocytosis regulators, including syntaxin, synapsin, RIM and Ca2+ channels. Moreover, calmodulin interacts with synaptotagmin through either direct binding or indirect phosphorylation. In summary, calmodulin and synaptotagmin are Ca2+ sensors that play complementary roles throughout the process of exocytosis. In this review, we discuss the complementary roles that calmodulin and synaptotagmin play as Ca2+ sensors during exocytosis.
Collapse
Affiliation(s)
- Renhao Xue
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Hao Meng
- Guangzhou Laboratory, Guangzhou, China.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Jiaxiang Yin
- Guangzhou Laboratory, Guangzhou, China.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Jingyao Xia
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Zhitao Hu
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Huisheng Liu
- Guangzhou Laboratory, Guangzhou, China.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| |
Collapse
|
31
|
Wang QW, Wang YH, Wang B, Chen Y, Lu SY, Yao J. Synaptotagmin-7-mediated activation of spontaneous NMDAR currents is disrupted in bipolar disorder susceptibility variants. PLoS Biol 2021; 19:e3001323. [PMID: 34228711 PMCID: PMC8284830 DOI: 10.1371/journal.pbio.3001323] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 07/16/2021] [Accepted: 06/15/2021] [Indexed: 11/19/2022] Open
Abstract
Synaptotagmin-7 (Syt7) plays direct or redundant Ca2+ sensor roles in multiple forms of vesicle exocytosis in synapses. Here, we show that Syt7 is a redundant Ca2+ sensor with Syt1/Doc2 to drive spontaneous glutamate release, which functions uniquely to activate the postsynaptic GluN2B-containing NMDARs that significantly contribute to mental illness. In mouse hippocampal neurons lacking Syt1/Doc2, Syt7 inactivation largely diminishes spontaneous release. Using 2 approaches, including measuring Ca2+ dose response and substituting extracellular Ca2+ with Sr2+, we detect that Syt7 directly triggers spontaneous release via its Ca2+ binding motif to activate GluN2B-NMDARs. Furthermore, modifying the localization of Syt7 in the active zone still allows Syt7 to drive spontaneous release, but the GluN2B-NMDAR activity is abolished. Finally, Syt7 SNPs identified in bipolar disorder patients destroy the function of Syt7 in spontaneous release in patient iPSC-derived and mouse hippocampal neurons. Therefore, Syt7 could contribute to neuropsychiatric disorders through driving spontaneous glutamate release.
Collapse
Affiliation(s)
- Qiu-Wen Wang
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ying-Han Wang
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Bing Wang
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yun Chen
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Si-Yao Lu
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jun Yao
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
32
|
van Westen R, Poppinga J, Díez Arazola R, Toonen RF, Verhage M. Neuromodulator release in neurons requires two functionally redundant calcium sensors. Proc Natl Acad Sci U S A 2021; 118:e2012137118. [PMID: 33903230 PMCID: PMC8106342 DOI: 10.1073/pnas.2012137118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neuropeptides and neurotrophic factors secreted from dense core vesicles (DCVs) control many brain functions, but the calcium sensors that trigger their secretion remain unknown. Here, we show that in mouse hippocampal neurons, DCV fusion is strongly and equally reduced in synaptotagmin-1 (Syt1)- or Syt7-deficient neurons, but combined Syt1/Syt7 deficiency did not reduce fusion further. Cross-rescue, expression of Syt1 in Syt7-deficient neurons, or vice versa, completely restored fusion. Hence, both sensors are rate limiting, operating in a single pathway. Overexpression of either sensor in wild-type neurons confirmed this and increased fusion. Syt1 traveled with DCVs and was present on fusing DCVs, but Syt7 supported fusion largely from other locations. Finally, the duration of single DCV fusion events was reduced in Syt1-deficient but not Syt7-deficient neurons. In conclusion, two functionally redundant calcium sensors drive neuromodulator secretion in an expression-dependent manner. In addition, Syt1 has a unique role in regulating fusion pore duration.
Collapse
Affiliation(s)
- Rhodé van Westen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Clinical Genetics, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
| | - Josse Poppinga
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Rocío Díez Arazola
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Ruud F Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
- Department of Clinical Genetics, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
33
|
Bourgeois-Jaarsma Q, Miaja Hernandez P, Groffen AJ. Ca 2+ sensor proteins in spontaneous release and synaptic plasticity: Limited contribution of Doc2c, rabphilin-3a and synaptotagmin 7 in hippocampal glutamatergic neurons. Mol Cell Neurosci 2021; 112:103613. [PMID: 33753311 DOI: 10.1016/j.mcn.2021.103613] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 03/09/2021] [Accepted: 03/13/2021] [Indexed: 11/28/2022] Open
Abstract
Presynaptic neurotransmitter release is strictly regulated by SNARE proteins, Ca2+ and a number of Ca2+ sensors including synaptotagmins (Syts) and Double C2 domain proteins (Doc2s). More than seventy years after the original description of spontaneous release, the mechanism that regulates this process is still poorly understood. Syt-1, Syt7 and Doc2 proteins contribute predominantly, but not exclusively, to synchronous, asynchronous and spontaneous phases of release. The proteins share a conserved tandem C2 domain architecture, but are functionally diverse in their subcellular location, Ca2+-binding properties and protein interactions. In absence of Syt-1, Doc2a and -b, neurons still exhibit spontaneous vesicle fusion which remains Ca2+-sensitive, suggesting the existence of additional sensors. Here, we selected Doc2c, rabphilin-3a and Syt-7 as three potential Ca2+ sensors for their sequence homology with Syt-1 and Doc2b. We genetically ablated each candidate gene in absence of Doc2a and -b and investigated spontaneous and evoked release in glutamatergic hippocampal neurons, cultured either in networks or on microglial islands (autapses). The removal of Doc2c had no effect on spontaneous or evoked release. Syt-7 removal also did not affect spontaneous release, although it altered short-term plasticity by accentuating short-term depression. The removal of rabphilin caused an increased spontaneous release frequency in network cultures, an effect that was not observed in autapses. Taken together, we conclude that Doc2c and Syt-7 do not affect spontaneous release of glutamate in hippocampal neurons, while our results suggest a possible regulatory role of rabphilin-3a in neuronal networks. These findings importantly narrow down the repertoire of synaptic Ca2+ sensors that may be implicated in the spontaneous release of glutamate.
Collapse
Affiliation(s)
- Quentin Bourgeois-Jaarsma
- Department of Functional Genomics, Faculty of Science, Center for Neurogenomics and Cognitive Research, VU University, De Boelelaan 1085, 1081HV Amsterdam, the Netherlands
| | - Pablo Miaja Hernandez
- Department of Functional Genomics, Faculty of Science, Center for Neurogenomics and Cognitive Research, VU University, De Boelelaan 1085, 1081HV Amsterdam, the Netherlands
| | - Alexander J Groffen
- Department of Functional Genomics, Faculty of Science, Center for Neurogenomics and Cognitive Research, VU University, De Boelelaan 1085, 1081HV Amsterdam, the Netherlands; Department of Clinical Genetics, VU Medical Center, De Boelelaan 1085, 1081HV Amsterdam, the Netherlands.
| |
Collapse
|
34
|
Abramov D, Guiberson NGL, Burré J. STXBP1 encephalopathies: Clinical spectrum, disease mechanisms, and therapeutic strategies. J Neurochem 2021; 157:165-178. [PMID: 32643187 PMCID: PMC7812771 DOI: 10.1111/jnc.15120] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/24/2020] [Accepted: 06/30/2020] [Indexed: 12/13/2022]
Abstract
Mutations in Munc18-1/STXBP1 (syntaxin-binding protein 1) are linked to various severe early epileptic encephalopathies and neurodevelopmental disorders. Heterozygous mutations in the STXBP1 gene include missense, nonsense, frameshift, and splice site mutations, as well as intragenic deletions and duplications and whole-gene deletions. No genotype-phenotype correlation has been identified so far, and patients are treated by anti-epileptic drugs because of the lack of a specific disease-modifying therapy. The molecular disease mechanisms underlying STXBP1-linked disorders are yet to be fully understood, but both haploinsufficiency and dominant-negative mechanisms have been proposed. This review focuses on the current understanding of the phenotypic spectrum of STXBP1-linked disorders, as well as discusses disease mechanisms in the context of the numerous pathways in which STXBP1 functions in the brain. We additionally evaluate the available animal models to study these disorders and highlight potential therapeutic approaches for treating these devastating diseases.
Collapse
Affiliation(s)
- Debra Abramov
- Appel Institute for Alzheimer's Disease Research, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Noah Guy Lewis Guiberson
- Appel Institute for Alzheimer's Disease Research, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Jacqueline Burré
- Appel Institute for Alzheimer's Disease Research, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
35
|
Li J, Deng S, He Q, Ke W, Shu Y. Asynchronous Glutamate Release at Autapses Regulates Spike Reliability and Precision in Mouse Neocortical Pyramidal Cells. Cereb Cortex 2021; 31:2278-2290. [PMID: 33279969 DOI: 10.1093/cercor/bhaa361] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 11/13/2022] Open
Abstract
Autapses are self-synapses of a neuron. Inhibitory autapses in the neocortex release GABA in 2 modes, synchronous release and asynchronous release (AR), providing precise and prolonged self-inhibition, respectively. A subpopulation of neocortical pyramidal cells (PCs) also forms functional autapses, activation of which promotes burst firing by strong unitary autaptic response that reflects synchronous glutamate release. However, it remains unclear whether AR occurs at PC autapses and plays a role in neuronal signaling. We performed whole-cell recordings from layer-5 PCs in slices of mouse prefrontal cortex (PFC). In response to action potential (AP) burst, 63% of PCs showed robust long-lasting autaptic AR, much stronger than synaptic AR between neighboring PCs. The autaptic AR is mediated predominantly by P/Q-type Ca2+ channels, and its strength depends on the intensity of PC activity and the level of residual Ca2+. Further experiments revealed that autaptic AR enhances spiking activities but reduces the temporal precision of post-burst APs. Together, the results show the occurrence of AR at PC autapses, the delayed and persistent glutamate AR causes self-excitation in individual PCs but may desynchronize the autaptic PC population. Thus, glutamatergic autapses should be essential elements in PFC and contribute to cortical information processing.
Collapse
Affiliation(s)
- Junlong Li
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Suixin Deng
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Quansheng He
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Wei Ke
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yousheng Shu
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
36
|
Function of Drosophila Synaptotagmins in membrane trafficking at synapses. Cell Mol Life Sci 2021; 78:4335-4364. [PMID: 33619613 PMCID: PMC8164606 DOI: 10.1007/s00018-021-03788-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/29/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022]
Abstract
The Synaptotagmin (SYT) family of proteins play key roles in regulating membrane trafficking at neuronal synapses. Using both Ca2+-dependent and Ca2+-independent interactions, several SYT isoforms participate in synchronous and asynchronous fusion of synaptic vesicles (SVs) while preventing spontaneous release that occurs in the absence of stimulation. Changes in the function or abundance of the SYT1 and SYT7 isoforms alter the number and route by which SVs fuse at nerve terminals. Several SYT family members also regulate trafficking of other subcellular organelles at synapses, including dense core vesicles (DCV), exosomes, and postsynaptic vesicles. Although SYTs are linked to trafficking of multiple classes of synaptic membrane compartments, how and when they interact with lipids, the SNARE machinery and other release effectors are still being elucidated. Given mutations in the SYT family cause disorders in both the central and peripheral nervous system in humans, ongoing efforts are defining how these proteins regulate vesicle trafficking within distinct neuronal compartments. Here, we review the Drosophila SYT family and examine their role in synaptic communication. Studies in this invertebrate model have revealed key similarities and several differences with the predicted activity of their mammalian counterparts. In addition, we highlight the remaining areas of uncertainty in the field and describe outstanding questions on how the SYT family regulates membrane trafficking at nerve terminals.
Collapse
|
37
|
朱 佳. Effects of Tetrodotoxin on Different Types of Synaptic Secretion. Biophysics (Nagoya-shi) 2021. [DOI: 10.12677/biphy.2021.93007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
38
|
Mochida S. Neurotransmitter Release Site Replenishment and Presynaptic Plasticity. Int J Mol Sci 2020; 22:ijms22010327. [PMID: 33396919 PMCID: PMC7794938 DOI: 10.3390/ijms22010327] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 12/23/2020] [Accepted: 12/27/2020] [Indexed: 12/19/2022] Open
Abstract
An action potential (AP) triggers neurotransmitter release from synaptic vesicles (SVs) docking to a specialized release site of presynaptic plasma membrane, the active zone (AZ). The AP simultaneously controls the release site replenishment with SV for sustainable synaptic transmission in response to incoming neuronal signals. Although many studies have suggested that the replenishment time is relatively slow, recent studies exploring high speed resolution have revealed SV dynamics with milliseconds timescale after an AP. Accurate regulation is conferred by proteins sensing Ca2+ entering through voltage-gated Ca2+ channels opened by an AP. This review summarizes how millisecond Ca2+ dynamics activate multiple protein cascades for control of the release site replenishment with release-ready SVs that underlie presynaptic short-term plasticity.
Collapse
Affiliation(s)
- Sumiko Mochida
- Department of Physiology, Tokyo Medical University, Tokyo 160-8402, Japan
| |
Collapse
|
39
|
Wang QW, Lu SY, Liu YN, Chen Y, Wei H, Shen W, Chen YF, Fu CL, Wang YH, Dai A, Huang X, Gage FH, Xu Q, Yao J. Synaptotagmin-7 deficiency induces mania-like behavioral abnormalities through attenuating GluN2B activity. Proc Natl Acad Sci U S A 2020; 117:31438-31447. [PMID: 33229564 PMCID: PMC7733786 DOI: 10.1073/pnas.2016416117] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Synaptotagmin-7 (Syt7) probably plays an important role in bipolar-like behavioral abnormalities in mice; however, the underlying mechanisms for this have remained elusive. Unlike antidepressants that cause mood overcorrection in bipolar depression, N-methyl-d-aspartate receptor (NMDAR)-targeted drugs show moderate clinical efficacy, for unexplained reasons. Here we identified Syt7 single nucleotide polymorphisms (SNPs) in patients with bipolar disorder and demonstrated that mice lacking Syt7 or expressing the SNPs showed GluN2B-NMDAR dysfunction, leading to antidepressant behavioral consequences and avoidance of overcorrection by NMDAR antagonists. In human induced pluripotent stem cell (iPSC)-derived and mouse hippocampal neurons, Syt7 and GluN2B-NMDARs were localized to the peripheral synaptic region, and Syt7 triggered multiple forms of glutamate release to efficiently activate the juxtaposed GluN2B-NMDARs. Thus, while Syt7 deficiency and SNPs induced GluN2B-NMDAR dysfunction in mice, patient iPSC-derived neurons showed Syt7 deficit-induced GluN2B-NMDAR hypoactivity that was rescued by Syt7 overexpression. Therefore, Syt7 deficits induced mania-like behaviors in mice by attenuating GluN2B activity, which enabled NMDAR antagonists to avoid mood overcorrection.
Collapse
Affiliation(s)
- Qiu-Wen Wang
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Si-Yao Lu
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Yao-Nan Liu
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Yun Chen
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Hui Wei
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences and Peking Union Medical College, Neuroscience Center, Chinese Academy of Medical Sciences, 100005 Beijing, China
| | - Wei Shen
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Yan-Fen Chen
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Chong-Lei Fu
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Ying-Han Wang
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Anbang Dai
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Xuan Huang
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, 100020 Beijing, China
| | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Qi Xu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences and Peking Union Medical College, Neuroscience Center, Chinese Academy of Medical Sciences, 100005 Beijing, China;
| | - Jun Yao
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China;
| |
Collapse
|
40
|
Yusipov I, Bacalini MG, Kalyakulina A, Krivonosov M, Pirazzini C, Gensous N, Ravaioli F, Milazzo M, Giuliani C, Vedunova M, Fiorito G, Gagliardi A, Polidoro S, Garagnani P, Ivanchenko M, Franceschi C. Age-related DNA methylation changes are sex-specific: a comprehensive assessment. Aging (Albany NY) 2020; 12:24057-24080. [PMID: 33276343 PMCID: PMC7762479 DOI: 10.18632/aging.202251] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/19/2020] [Indexed: 12/18/2022]
Abstract
The existence of a sex gap in human health and longevity has been widely documented. Autosomal DNA methylation differences between males and females have been reported, but so far few studies have investigated if DNA methylation is differently affected by aging in males and females. We performed a meta-analysis of 4 large whole blood datasets, comparing 4 aspects of epigenetic age-dependent remodeling between the two sexes: differential methylation, variability, epimutations and entropy. We reported that a large fraction (43%) of sex-associated probes undergoes age-associated DNA methylation changes, and that a limited number of probes show age-by-sex interaction. We experimentally validated 2 regions mapping in FIGN and PRR4 genes and showed sex-specific deviations of their methylation patterns in models of decelerated (centenarians) and accelerated (Down syndrome) aging. While we did not find sex differences in the age-associated increase in epimutations and entropy, we showed that the number of probes having an age-related increase in methylation variability is 15 times higher in males compared to females. Our results can offer new epigenetic tools to study the interaction between aging and sex and can pave the way to the identification of molecular triggers of sex differences in longevity and age-related diseases prevalence.
Collapse
Affiliation(s)
- Igor Yusipov
- Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky University, Nizhniy Novgorod, Russia.,Mathematics of Future Technologies Center, Lobachevsky University, Nizhniy Novgorod, Russia
| | | | - Alena Kalyakulina
- Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky University, Nizhniy Novgorod, Russia
| | - Mikhail Krivonosov
- Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky University, Nizhniy Novgorod, Russia
| | - Chiara Pirazzini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Noémie Gensous
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum – University of Bologna, Bologna, Italy
| | - Francesco Ravaioli
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum – University of Bologna, Bologna, Italy
| | - Maddalena Milazzo
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum – University of Bologna, Bologna, Italy
| | - Cristina Giuliani
- Department of Biological, Geological, and Environmental Sciences (BiGeA), Laboratory of Molecular Anthropology and Centre for Genome Biology, University of Bologna, Bologna, Italy.,School of Anthropology and Museum Ethnography, University of Oxford, Oxford, UK
| | - Maria Vedunova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, Nizhni Novgorod, Russia
| | - Giovanni Fiorito
- Department of Biomedical Sciences, University of Sassari, Italy.,Department of Epidemiology and Public Health, MRC/HPA Centre for Environment and Health, School of Public Health, Imperial College London, Norfolk Place, London W2 1PG, UK
| | - Amedeo Gagliardi
- Italian Institute for Genomic Medicine (IIGM), Candiolo 10060, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo 10060, Italy
| | - Silvia Polidoro
- Department of Epidemiology and Public Health, MRC/HPA Centre for Environment and Health, School of Public Health, Imperial College London, Norfolk Place, London W2 1PG, UK.,Italian Institute for Genomic Medicine (IIGM), Candiolo 10060, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo 10060, Italy
| | - Paolo Garagnani
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum – University of Bologna, Bologna, Italy.,Department of Laboratory Medicine, Clinical Chemistry, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Applied Biomedical Research Center (CRBA), Policlinico S.Orsola-Malpighi Polyclinic, Bologna, Italy.,CNR Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy
| | - Mikhail Ivanchenko
- Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky University, Nizhniy Novgorod, Russia.,Mathematics of Future Technologies Center, Lobachevsky University, Nizhniy Novgorod, Russia
| | - Claudio Franceschi
- Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky University, Nizhniy Novgorod, Russia
| |
Collapse
|
41
|
Synaptic vesicles transiently dock to refill release sites. Nat Neurosci 2020; 23:1329-1338. [PMID: 32989294 PMCID: PMC8054220 DOI: 10.1038/s41593-020-00716-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 09/01/2020] [Indexed: 11/18/2022]
Abstract
Synaptic vesicles fuse with the plasma membrane to release neurotransmitter following an action potential, after which new vesicles must ‘dock’ to refill vacated release sites. To capture synaptic vesicle exocytosis at cultured mouse hippocampal synapses, we induced single action potentials by electrical field stimulation then subjected neurons to high-pressure freezing to examine their morphology by electron microscopy. During synchronous release, multiple vesicles can fuse at a single active zone. Fusions during synchronous release are distributed throughout the active zone, whereas fusions during asynchronous release are biased toward the center of the active zone. After stimulation, the total number of docked vesicles across all synapses decreases by ~40%. Within 14 ms, new vesicles are recruited and fully replenish the docked pool, but this docking is transient and they either undock or fuse within 100 ms. These results demonstrate that recruitment of synaptic vesicles to release sites is rapid and reversible.
Collapse
|
42
|
Bigan E, Sasidharan Nair S, Lejeune FX, Fragnaud H, Parmentier F, Mégret L, Verny M, Aaronson J, Rosinski J, Neri C. Genetic cooperativity in multi-layer networks implicates cell survival and senescence in the striatum of Huntington's disease mice synchronous to symptoms. Bioinformatics 2020; 36:186-196. [PMID: 31228193 PMCID: PMC6956776 DOI: 10.1093/bioinformatics/btz514] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 06/11/2019] [Accepted: 06/18/2019] [Indexed: 02/06/2023] Open
Abstract
Motivation Huntington’s disease (HD) may evolve through gene deregulation. However, the impact of gene deregulation on the dynamics of genetic cooperativity in HD remains poorly understood. Here, we built a multi-layer network model of temporal dynamics of genetic cooperativity in the brain of HD knock-in mice (allelic series of Hdh mice). To enhance biological precision and gene prioritization, we integrated three complementary families of source networks, all inferred from the same RNA-seq time series data in Hdh mice, into weighted-edge networks where an edge recapitulates path-length variation across source-networks and age-points. Results Weighted edge networks identify two consecutive waves of tight genetic cooperativity enriched in deregulated genes (critical phases), pre-symptomatically in the cortex, implicating neurotransmission, and symptomatically in the striatum, implicating cell survival (e.g. Hipk4) intertwined with cell proliferation (e.g. Scn4b) and cellular senescence (e.g. Cdkn2a products) responses. Top striatal weighted edges are enriched in modulators of defective behavior in invertebrate models of HD pathogenesis, validating their relevance to neuronal dysfunction in vivo. Collectively, these findings reveal highly dynamic temporal features of genetic cooperativity in the brain of Hdh mice where a 2-step logic highlights the importance of cellular maintenance and senescence in the striatum of symptomatic mice, providing highly prioritized targets. Availability and implementation Weighted edge network analysis (WENA) data and source codes for performing spectral decomposition of the signal (SDS) and WENA analysis, both written using Python, are available at http://www.broca.inserm.fr/HD-WENA/. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Erwan Bigan
- Sorbonnes Université, Centre National de la Recherche Scientifique, Research Unit Biology of Adaptation and Aging (B2A), Team Compensation in Neurodegenerative Diseases and Aging (Brain-C), Paris F-75252, France
| | - Satish Sasidharan Nair
- Sorbonnes Université, Centre National de la Recherche Scientifique, Research Unit Biology of Adaptation and Aging (B2A), Team Compensation in Neurodegenerative Diseases and Aging (Brain-C), Paris F-75252, France
| | - François-Xavier Lejeune
- Sorbonnes Université, Centre National de la Recherche Scientifique, Research Unit Biology of Adaptation and Aging (B2A), Team Compensation in Neurodegenerative Diseases and Aging (Brain-C), Paris F-75252, France
| | - Hélissande Fragnaud
- Sorbonnes Université, Centre National de la Recherche Scientifique, Research Unit Biology of Adaptation and Aging (B2A), Team Compensation in Neurodegenerative Diseases and Aging (Brain-C), Paris F-75252, France
| | - Frédéric Parmentier
- Sorbonnes Université, Centre National de la Recherche Scientifique, Research Unit Biology of Adaptation and Aging (B2A), Team Compensation in Neurodegenerative Diseases and Aging (Brain-C), Paris F-75252, France
| | - Lucile Mégret
- Sorbonnes Université, Centre National de la Recherche Scientifique, Research Unit Biology of Adaptation and Aging (B2A), Team Compensation in Neurodegenerative Diseases and Aging (Brain-C), Paris F-75252, France
| | - Marc Verny
- Sorbonnes Université, Centre National de la Recherche Scientifique, Research Unit Biology of Adaptation and Aging (B2A), Team Compensation in Neurodegenerative Diseases and Aging (Brain-C), Paris F-75252, France
| | | | | | - Christian Neri
- Sorbonnes Université, Centre National de la Recherche Scientifique, Research Unit Biology of Adaptation and Aging (B2A), Team Compensation in Neurodegenerative Diseases and Aging (Brain-C), Paris F-75252, France
| |
Collapse
|
43
|
Bisio H, Soldati-Favre D. Signaling Cascades Governing Entry into and Exit from Host Cells by Toxoplasma gondii. Annu Rev Microbiol 2020; 73:579-599. [PMID: 31500539 DOI: 10.1146/annurev-micro-020518-120235] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The Apicomplexa phylum includes a large group of obligate intracellular protozoan parasites responsible for important diseases in humans and animals. Toxoplasma gondii is a widespread parasite with considerable versatility, and it is capable of infecting virtually any warm-blooded animal, including humans. This outstanding success can be attributed at least in part to an efficient and continuous sensing of the environment, with a ready-to-adapt strategy. This review updates the current understanding of the signals governing the lytic cycle of T. gondii, with particular focus on egress from infected cells, a key step for balancing survival, multiplication, and spreading in the host. We cover the recent advances in the conceptual framework of regulation of microneme exocytosis that ensures egress, motility, and invasion. Particular emphasis is given to the trigger molecules and signaling cascades regulating exit from host cells.
Collapse
Affiliation(s)
- Hugo Bisio
- Département de Microbiologie et Médecine Moléculaire, Centre Médical Universitaire, Université de Genève, 1211 Geneva 4, Switzerland;
| | - Dominique Soldati-Favre
- Département de Microbiologie et Médecine Moléculaire, Centre Médical Universitaire, Université de Genève, 1211 Geneva 4, Switzerland;
| |
Collapse
|
44
|
The Synaptic Vesicle Cycle Revisited: New Insights into the Modes and Mechanisms. J Neurosci 2020; 39:8209-8216. [PMID: 31619489 DOI: 10.1523/jneurosci.1158-19.2019] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/31/2019] [Accepted: 08/03/2019] [Indexed: 02/01/2023] Open
Abstract
Neurotransmission is sustained by endocytosis and refilling of synaptic vesicles (SVs) locally within the presynapse. Until recently, a consensus formed that after exocytosis, SVs are recovered by either fusion pore closure (kiss-and-run) or clathrin-mediated endocytosis directly from the plasma membrane. However, recent data have revealed that SV formation is more complex than previously envisaged. For example, two additional recycling pathways have been discovered, ultrafast endocytosis and activity-dependent bulk endocytosis, in which SVs are regenerated from the internalized membrane and synaptic endosomes. Furthermore, these diverse modes of endocytosis appear to influence both the molecular composition and subsequent physiological role of individual SVs. In addition, previously unknown complexity in SV refilling and reclustering has been revealed. This review presents a modern view of the SV life cycle and discusses how neuronal subtype, physiological temperature, and individual activity patterns can recruit different endocytic modes to generate new SVs and sculpt subsequent presynaptic performance.
Collapse
|
45
|
VAMP4 Maintains a Ca 2+-Sensitive Pool of Spontaneously Recycling Synaptic Vesicles. J Neurosci 2020; 40:5389-5401. [PMID: 32532887 DOI: 10.1523/jneurosci.2386-19.2020] [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: 10/02/2019] [Revised: 05/29/2020] [Accepted: 06/05/2020] [Indexed: 11/21/2022] Open
Abstract
Spontaneous neurotransmitter release is a fundamental property of synapses in which neurotransmitter filled vesicles release their content independent of presynaptic action potentials (APs). Despite their seemingly random nature, these spontaneous fusion events can be regulated by Ca2+ signaling pathways. Here, we probed the mechanisms that maintain Ca2+ sensitivity of spontaneous release events in synapses formed between hippocampal neurons cultured from rats of both sexes. In this setting, we examined the potential role of vesicle-associated membrane protein 4 (VAMP4), a vesicular soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein in spontaneous neurotransmission. Our results show that VAMP4 is required for Ca2+-dependent spontaneous excitatory neurotransmission, with a limited role in spontaneous inhibitory neurotransmission. Key residues in VAMP4 that regulate its retrieval as well as functional clathrin-mediated vesicle trafficking were essential for the maintenance of VAMP4-mediated spontaneous release. Moreover, high-frequency stimulation (HFS) that typically triggers asynchronous release and retrieval of VAMP4 from the plasma membrane also augmentsCa2+-sensitive spontaneous release for up to 30 min in a VAMP4-dependent manner. This VAMP4-mediated link between asynchronous and spontaneous excitatory neurotransmission might serve as a presynaptic substrate for synaptic plasticity coupling distinct forms of release.SIGNIFICANCE STATEMENT Spontaneous neurotransmitter release that occurs independent of presynaptic action potentials (APs) shows significant sensitivity to intracellular Ca2+ levels. In this study, we identify the vesicular soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) molecule vesicle-associated membrane protein 4 (VAMP4) as a key component of the machinery that maintains these Ca2+-sensitive fraction of spontaneous release events. Following brief intense activity, VAMP4-dependent synaptic vesicle retrieval supports a pool of vesicles that fuse spontaneously in the long term. We propose that this vesicle trafficking pathway acts to shape spontaneous release and associated signaling based on previous activity history of synapses.
Collapse
|
46
|
Banerjee A, Lee J, Nemcova P, Liu C, Kaeser PS. Synaptotagmin-1 is the Ca 2+ sensor for fast striatal dopamine release. eLife 2020; 9:58359. [PMID: 32490813 PMCID: PMC7319770 DOI: 10.7554/elife.58359] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/02/2020] [Indexed: 11/13/2022] Open
Abstract
Dopamine powerfully controls neural circuits through neuromodulation. In the vertebrate striatum, dopamine adjusts cellular functions to regulate behaviors across broad time scales, but how the dopamine secretory system is built to support fast and slow neuromodulation is not known. Here, we set out to identify Ca2+-triggering mechanisms for dopamine release. We find that synchronous dopamine secretion is abolished in acute brain slices of conditional knockout mice in which Synaptotagmin-1 is removed from dopamine neurons. This indicates that Synaptotagmin-1 is the Ca2+ sensor for fast dopamine release. Remarkably, dopamine release induced by strong depolarization and asynchronous release during stimulus trains are unaffected by Synaptotagmin-1 knockout. Microdialysis further reveals that these modes and action potential-independent release provide significant amounts of extracellular dopamine in vivo. We propose that the molecular machinery for dopamine secretion has evolved to support fast and slow signaling modes, with fast release requiring the Ca2+ sensor Synaptotagmin-1.
Collapse
Affiliation(s)
- Aditi Banerjee
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Jinoh Lee
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Paulina Nemcova
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Changliang Liu
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, United States
| |
Collapse
|
47
|
Khan MM, Regehr WG. Loss of Doc2b does not influence transmission at Purkinje cell to deep nuclei synapses under physiological conditions. eLife 2020; 9:55165. [PMID: 32347796 PMCID: PMC7190354 DOI: 10.7554/elife.55165] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/09/2020] [Indexed: 12/26/2022] Open
Abstract
Doc2a and Doc2b are high-affinity calcium-binding proteins that interact with SNARE proteins and phospholipids. Experiments performed on cultured cells indicated that Doc2 proteins promote spontaneous vesicle fusion and asynchronous neurotransmitter release, regulate vesicle priming, mediate augmentation, and regulate transmission during sustained activity. Here, we assess the role of Doc2 proteins in synaptic transmission under physiological conditions at mature synapses made by Purkinje cells onto neurons in the deep cerebellar nuclei (PC to DCN synapses). PCs express Doc2b but not Doc2a. Surprisingly, spontaneous neurotransmitter release, synaptic strength, the time course of evoked release, responses evoked by sustained high-frequency stimulation, and short-term plasticity were normal in Doc2b KO mice. Thus, in stark contrast to numerous functions previously proposed for Doc2, here we find that Doc2b removal does not influence transmission at PC-to-DCN synapses, indicating that conclusions based on studies of Doc2b in cultured cells do not necessarily generalize to mature synapses under physiological conditions.
Collapse
Affiliation(s)
- Mehak M Khan
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Wade G Regehr
- Department of Neurobiology, Harvard Medical School, Boston, United States
| |
Collapse
|
48
|
Doc2 Proteins Are Not Required for the Increased Spontaneous Release Rate in Synaptotagmin-1-Deficient Neurons. J Neurosci 2020; 40:2606-2617. [PMID: 32098902 DOI: 10.1523/jneurosci.0309-19.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/10/2020] [Accepted: 01/23/2020] [Indexed: 11/21/2022] Open
Abstract
Regulated secretion is controlled by Ca2+ sensors with different affinities and subcellular distributions. Inactivation of Syt1 (synaptotagmin-1), the main Ca2+ sensor for synchronous neurotransmission in many neurons, enhances asynchronous and spontaneous release rates, suggesting that Syt1 inhibits other sensors with higher Ca2+ affinities and/or lower cooperativities. Such sensors could include Doc2a and Doc2b, which have been implicated in spontaneous and asynchronous neurotransmitter release and compete with Syt1 for binding SNARE complexes. Here, we tested this hypothesis using triple-knock-out mice. Inactivation of Doc2a and Doc2b in Syt1-deficient neurons did not reduce the high spontaneous release rate. Overexpression of Doc2b variants in triple-knock-out neurons reduced spontaneous release but did not rescue synchronous release. A chimeric construct in which the C2AB domain of Syt1 was substituted by that of Doc2b did not support synchronous release either. Conversely, the soluble C2AB domain of Syt1 did not affect spontaneous release. We conclude that the high spontaneous release rate in synaptotagmin-deficient neurons does not involve the binding of Doc2 proteins to Syt1 binding sites in the SNARE complex. Instead, our results suggest that the C2AB domains of Syt1 and Doc2b specifically support synchronous and spontaneous release by separate mechanisms. (Both male and female neurons were studied without sex determination.)SIGNIFICANCE STATEMENT Neurotransmission in the brain is regulated by presynaptic Ca2+ concentrations. Multiple Ca2+ sensor proteins contribute to synchronous (Syt1, Syt2), asynchronous (Syt7), and spontaneous (Doc2a/Doc2b) phases of neurotransmitter release. Genetic ablation of synchronous release was previously shown to affect other release phases, suggesting that multiple sensors may compete for similar release sites, together encoding stimulus-secretion coupling over a large range of synaptic Ca2+ concentrations. Here, we investigated the extent of functional overlap between Syt1, Doc2a, and Doc2b by reintroducing wild-type and mutant proteins in triple-knock-out neurons, and conclude that the sensors are highly specialized for different phases of release.
Collapse
|
49
|
Regulation of Recurrent Inhibition by Asynchronous Glutamate Release in Neocortex. Neuron 2020; 105:522-533.e4. [DOI: 10.1016/j.neuron.2019.10.038] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 06/11/2019] [Accepted: 10/28/2019] [Indexed: 01/06/2023]
|
50
|
Akula AK, Zhang X, Viotti JS, Nestvogel D, Rhee JS, Ebrecht R, Reim K, Wouters F, Liepold T, Jahn O, Bogeski I, Dresbach T. The Calmodulin Binding Region of the Synaptic Vesicle Protein Mover Is Required for Homomeric Interaction and Presynaptic Targeting. Front Mol Neurosci 2019; 12:249. [PMID: 31787876 PMCID: PMC6856015 DOI: 10.3389/fnmol.2019.00249] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 09/26/2019] [Indexed: 01/10/2023] Open
Abstract
Neurotransmitter release is mediated by an evolutionarily conserved machinery. The synaptic vesicle (SV) associated protein Mover/TPRGL/SVAP30 does not occur in all species and all synapses. Little is known about its molecular properties and how it may interact with the conserved components of the presynaptic machinery. Here, we show by deletion analysis that regions required for homomeric interaction of Mover are distributed across the entire molecule, including N-terminal, central and C-terminal regions. The same regions are also required for the accumulation of Mover in presynaptic terminals of cultured neurons. Mutating two phosphorylation sites in N-terminal regions did not affect these properties. In contrast, a point mutation in the predicted Calmodulin (CaM) binding sequence of Mover abolished both homomeric interaction and presynaptic targeting. We show that this sequence indeed binds Calmodulin, and that recombinant Mover increases Calmodulin signaling upon heterologous expression. Our data suggest that presynaptic accumulation of Mover requires homomeric interaction mediated by regions distributed across large areas of the protein, and corroborate the hypothesis that Mover functionally interacts with Calmodulin signaling.
Collapse
Affiliation(s)
- Asha Kiran Akula
- Institute for Anatomy and Embryology, University Medical Center Göttingen, Göttingen, Germany
| | - Xin Zhang
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Göttingen, Germany
| | - Julio S Viotti
- Institute for Anatomy and Embryology, University Medical Center Göttingen, Göttingen, Germany
| | - Dennis Nestvogel
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Jeong-Seop Rhee
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Rene Ebrecht
- Institute for Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Kerstin Reim
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Fred Wouters
- Institute for Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Thomas Liepold
- Proteomics Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Olaf Jahn
- Proteomics Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Ivan Bogeski
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Göttingen, Germany
| | - Thomas Dresbach
- Institute for Anatomy and Embryology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|