1
|
Trus M, Atlas D. Non-ionotropic voltage-gated calcium channel signaling. Channels (Austin) 2024; 18:2341077. [PMID: 38601983 PMCID: PMC11017947 DOI: 10.1080/19336950.2024.2341077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024] Open
Abstract
Voltage-gated calcium channels (VGCCs) are the major conduits for calcium ions (Ca2+) within excitable cells. Recent studies have highlighted the non-ionotropic functionality of VGCCs, revealing their capacity to activate intracellular pathways independently of ion flow. This non-ionotropic signaling mode plays a pivotal role in excitation-coupling processes, including gene transcription through excitation-transcription (ET), synaptic transmission via excitation-secretion (ES), and cardiac contraction through excitation-contraction (EC). However, it is noteworthy that these excitation-coupling processes require extracellular calcium (Ca2+) and Ca2+ occupancy of the channel ion pore. Analogous to the "non-canonical" characterization of the non-ionotropic signaling exhibited by the N-methyl-D-aspartate receptor (NMDA), which requires extracellular Ca2+ without the influx of ions, VGCC activation requires depolarization-triggered conformational change(s) concomitant with Ca2+ binding to the open channel. Here, we discuss the contributions of VGCCs to ES, ET, and EC coupling as Ca2+ binding macromolecules that transduces external stimuli to intracellular input prior to elevating intracellular Ca2+. We emphasize the recognition of calcium ion occupancy within the open ion-pore and its contribution to the excitation coupling processes that precede the influx of calcium. The non-ionotropic activation of VGCCs, triggered by the upstroke of an action potential, provides a conceptual framework to elucidate the mechanistic aspects underlying the microseconds nature of synaptic transmission, cardiac contractility, and the rapid induction of first-wave genes.
Collapse
Affiliation(s)
- Michael Trus
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Daphne Atlas
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
2
|
Li K, Li K, Fan J, Zhang X, Tao C, Xiang Y, Cui L, Li H, Li M, Zhang Y, Geng J, Lai Y. Synergistic regulation of fusion pore opening and dilation by SNARE and synaptotagmin-1. J Mol Cell Biol 2024; 16:mjae011. [PMID: 38444183 PMCID: PMC11472156 DOI: 10.1093/jmcb/mjae011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/19/2023] [Accepted: 03/04/2024] [Indexed: 03/07/2024] Open
Abstract
Fusion pore opening is a transient intermediate state of synaptic vesicle exocytosis, which is highly dynamic and precisely regulated by the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex and synaptotagmin-1 (Syt1). Yet, the regulatory mechanism is not fully understood. In this work, using single-channel membrane fusion electrophysiology, we determined that SNAREpins are important for driving fusion pore opening and dilation but incapable of regulating the dynamics. When Syt1 was added, the closing frequency of fusion pores significantly increased, while the radius of fusion pores mildly decreased. In response to Ca2+, SNARE/Syt1 greatly increased the radius of fusion pores and reduced their closing frequency. Moreover, the residue F349 in the C2B domain of Syt1, which mediates Syt1 oligomerization, was required for clamping fusion pore opening in the absence of Ca2+, probably by extending the distance between the two membranes. Finally, in Ca2+-triggered fusion, the primary interface between SNARE and Syt1 plays a critical role in stabilizing and dilating the fusion pore, while the polybasic region of Syt1 C2B domain has a mild effect on increasing the radius of the fusion pore. In summary, our results suggest that Syt1, SNARE, and the anionic membrane synergically orchestrate the dynamics of fusion pore opening in synaptic vesicle exocytosis.
Collapse
Affiliation(s)
- Kaiju Li
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Kaiyu Li
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiaqi Fan
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xing Zhang
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chengyan Tao
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Yijuan Xiang
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lele Cui
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hao Li
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Minghan Li
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Yanjing Zhang
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Jia Geng
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu 641400, China
| | - Ying Lai
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
3
|
Duan J, Kahms M, Steinhoff A, Klingauf J. Spontaneous and evoked synaptic vesicle release arises from a single releasable pool. Cell Rep 2024; 43:114461. [PMID: 38990719 DOI: 10.1016/j.celrep.2024.114461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 05/23/2024] [Accepted: 06/23/2024] [Indexed: 07/13/2024] Open
Abstract
The quantal content of an evoked postsynaptic response is typically determined by dividing it by the average spontaneous miniature response. However, this approach is challenged by the notion that different synaptic vesicle pools might drive spontaneous and evoked release. Here, we "silence" synaptic vesicles through pharmacological alkalinization and subsequently rescue them by optogenetic acidification. We find that such silenced synaptic vesicles, retrieved during evoked or spontaneous activity, cross-deplete the complementary release mode in a fully reversible manner. A fluorescently tagged version of the endosomal SNARE protein Vti1a, which has been suggested to identify a separate pool of spontaneously recycling synaptic vesicles, is trafficked to synaptic vesicles significantly only upon overexpression but not when endogenously tagged by CRISPR-Cas9. Thus, both release modes draw synaptic vesicles from the same readily releasable pool.
Collapse
Affiliation(s)
- Junxiu Duan
- Department of Cellular Biophysics, Institute of Medical Physics and Biophysics, University of Münster, Robert-Koch-Str. 31, 48149 Münster, Germany; Center for Soft Nanoscience SoN, University of Münster, Busso-Peus-Str.10, 48149 Münster, Germany; Cells in Motion Interfaculty Center, University of Münster, 48149 Münster, Germany; CiM Graduate School of the Cells in Motion Interfaculty Centre and the International Max Planck Research School, 48149 Münster, Germany
| | - Martin Kahms
- Department of Cellular Biophysics, Institute of Medical Physics and Biophysics, University of Münster, Robert-Koch-Str. 31, 48149 Münster, Germany; Center for Soft Nanoscience SoN, University of Münster, Busso-Peus-Str.10, 48149 Münster, Germany; Cells in Motion Interfaculty Center, University of Münster, 48149 Münster, Germany
| | - Ana Steinhoff
- Department of Cellular Biophysics, Institute of Medical Physics and Biophysics, University of Münster, Robert-Koch-Str. 31, 48149 Münster, Germany; Center for Soft Nanoscience SoN, University of Münster, Busso-Peus-Str.10, 48149 Münster, Germany; Cells in Motion Interfaculty Center, University of Münster, 48149 Münster, Germany; CiM Graduate School of the Cells in Motion Interfaculty Centre and the International Max Planck Research School, 48149 Münster, Germany
| | - Jürgen Klingauf
- Department of Cellular Biophysics, Institute of Medical Physics and Biophysics, University of Münster, Robert-Koch-Str. 31, 48149 Münster, Germany; Center for Soft Nanoscience SoN, University of Münster, Busso-Peus-Str.10, 48149 Münster, Germany; Cells in Motion Interfaculty Center, University of Münster, 48149 Münster, Germany.
| |
Collapse
|
4
|
van Boven MA, Mestroni M, Zwijnenburg PJG, Verhage M, Cornelisse LN. A de novo missense mutation in synaptotagmin-1 associated with neurodevelopmental disorder desynchronizes neurotransmitter release. Mol Psychiatry 2024; 29:1798-1809. [PMID: 38321119 PMCID: PMC11371641 DOI: 10.1038/s41380-024-02444-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/12/2024] [Accepted: 01/22/2024] [Indexed: 02/08/2024]
Abstract
Synaptotagmin-1 (Syt1) is a presynaptic calcium sensor with two calcium binding domains, C2A and C2B, that triggers action potential-induced synchronous neurotransmitter release, while suppressing asynchronous and spontaneous release. We identified a de novo missense mutation (P401L) in the C2B domain in a patient with developmental delay and autistic symptoms. Expressing the orthologous mouse mutant (P400L) in cultured Syt1 null mutant neurons revealed a reduction in dendrite outgrowth with a proportional reduction in synapses. This was not observed in single Syt1PL-rescued neurons that received normal synaptic input when cultured in a control network. Patch-clamp recordings showed that spontaneous miniature release events per synapse were increased more than 500% in Syt1PL-rescued neurons, even beyond the increased rates in Syt1 KO neurons. Furthermore, action potential-induced asynchronous release was increased more than 100%, while synchronous release was unaffected. A similar shift to more asynchronous release was observed during train stimulations. These cellular phenotypes were also observed when Syt1PL was overexpressed in wild type neurons. Our findings show that Syt1PL desynchronizes neurotransmission by increasing the readily releasable pool for asynchronous release and reducing the suppression of spontaneous and asynchronous release. Neurons respond to this by shortening their dendrites, possibly to counteract the increased synaptic input. Syt1PL acts in a dominant-negative manner supporting a causative role for the mutation in the heterozygous patient. We propose that the substitution of a rigid proline to a more flexible leucine at the bottom of the C2B domain impairs clamping of release by interfering with Syt1's primary interface with the SNARE complex. This is a novel cellular phenotype, distinct from what was previously found for other SYT1 disease variants, and points to a role for spontaneous and asynchronous release in SYT1-associated neurodevelopmental disorder.
Collapse
Affiliation(s)
- Maaike A van Boven
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Marta Mestroni
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | | | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) Amsterdam, 1081 HV, Amsterdam, The Netherlands
- Department of Functional Genomics and Department of Human Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam UMC-Location VUmc, 1081 HV, Amsterdam, The Netherlands
| | - L Niels Cornelisse
- Department of Functional Genomics and Department of Human Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam UMC-Location VUmc, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
5
|
Rademacher K, Doric Z, Haddad D, Mamaligas A, Liao SC, Creed RB, Kano K, Chatterton Z, Fu Y, Garcia JH, Vance V, Sei Y, Kreitzer A, Halliday GM, Nelson AB, Margolis EB, Nakamura K. Chronic hyperactivation of midbrain dopamine neurons causes preferential dopamine neuron degeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.05.588321. [PMID: 38645054 PMCID: PMC11030348 DOI: 10.1101/2024.04.05.588321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Parkinson's disease (PD) is characterized by the death of substantia nigra (SNc) dopamine (DA) neurons, but the pathophysiological mechanisms that precede and drive their death remain unknown. The activity of DA neurons is likely altered in PD, but we understand little about if or how chronic changes in activity may contribute to degeneration. To address this question, we developed a chemogenetic (DREADD) mouse model to chronically increase DA neuron activity, and confirmed this increase using ex vivo electrophysiology. Chronic hyperactivation of DA neurons resulted in prolonged increases in locomotor activity during the light cycle and decreases during the dark cycle, consistent with chronic changes in DA release and circadian disturbances. We also observed early, preferential degeneration of SNc projections, recapitulating the PD hallmarks of selective vulnerability of SNc axons and the comparative resilience of ventral tegmental area axons. This was followed by eventual loss of midbrain DA neurons. Continuous DREADD activation resulted in a sustained increase in baseline calcium levels, supporting an important role for increased calcium in the neurodegeneration process. Finally, spatial transcriptomics from DREADD mice examining midbrain DA neurons and striatal targets, and cross-validation with human patient samples, provided insights into potential mechanisms of hyperactivity-induced toxicity and PD. Our results thus reveal the preferential vulnerability of SNc DA neurons to increased neural activity, and support a potential role for increased neural activity in driving degeneration in PD.
Collapse
Affiliation(s)
- Katerina Rademacher
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Graduate Program in Neuroscience, University of California San Francisco, San Francisco , CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| | - Zak Doric
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Graduate Program in Neuroscience, University of California San Francisco, San Francisco , CA
| | - Dominik Haddad
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
| | - Aphroditi Mamaligas
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
| | - Szu-Chi Liao
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, CA
- Endocrinology Graduate Program, University of California Berkeley, Berkeley, CA
| | - Rose B. Creed
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA
| | - Kohei Kano
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| | - Zac Chatterton
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- Brain and Mind Centre, Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Yuhong Fu
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- Brain and Mind Centre, Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Joseph H. Garcia
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- School of Medicine, University of California, San Francisco, California, USA
| | - Victoria Vance
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- College of Science, Northeastern University, Boston, MA
| | - Yoshitaka Sei
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| | - Anatol Kreitzer
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Graduate Program in Neuroscience, University of California San Francisco, San Francisco , CA
- UCSF Department of Physiology, University of California San Francisco, CA
| | - Glenda M Halliday
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- Brain and Mind Centre, Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Alexandra B. Nelson
- Graduate Program in Neuroscience, University of California San Francisco, San Francisco , CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA
| | - Elyssa B. Margolis
- Graduate Program in Neuroscience, University of California San Francisco, San Francisco , CA
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA
| | - Ken Nakamura
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Graduate Program in Neuroscience, University of California San Francisco, San Francisco , CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA
- Graduate Program in Biomedical Sciences, University of California San Francisco, San Francisco
| |
Collapse
|
6
|
Kádková A, Murach J, Østergaard M, Malsam A, Malsam J, Lolicato F, Nickel W, Söllner TH, Sørensen JB. SNAP25 disease mutations change the energy landscape for synaptic exocytosis due to aberrant SNARE interactions. eLife 2024; 12:RP88619. [PMID: 38411501 PMCID: PMC10911398 DOI: 10.7554/elife.88619] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024] Open
Abstract
SNAP25 is one of three neuronal SNAREs driving synaptic vesicle exocytosis. We studied three mutations in SNAP25 that cause epileptic encephalopathy: V48F, and D166Y in the synaptotagmin-1 (Syt1)-binding interface, and I67N, which destabilizes the SNARE complex. All three mutations reduced Syt1-dependent vesicle docking to SNARE-carrying liposomes and Ca2+-stimulated membrane fusion in vitro and when expressed in mouse hippocampal neurons. The V48F and D166Y mutants (with potency D166Y > V48F) led to reduced readily releasable pool (RRP) size, due to increased spontaneous (miniature Excitatory Postsynaptic Current, mEPSC) release and decreased priming rates. These mutations lowered the energy barrier for fusion and increased the release probability, which are gain-of-function features not found in Syt1 knockout (KO) neurons; normalized mEPSC release rates were higher (potency D166Y > V48F) than in the Syt1 KO. These mutations (potency D166Y > V48F) increased spontaneous association to partner SNAREs, resulting in unregulated membrane fusion. In contrast, the I67N mutant decreased mEPSC frequency and evoked EPSC amplitudes due to an increase in the height of the energy barrier for fusion, whereas the RRP size was unaffected. This could be partly compensated by positive charges lowering the energy barrier. Overall, pathogenic mutations in SNAP25 cause complex changes in the energy landscape for priming and fusion.
Collapse
Affiliation(s)
- Anna Kádková
- Department of Neuroscience, University of CopenhagenCopenhagenDenmark
| | | | - Maiken Østergaard
- Department of Neuroscience, University of CopenhagenCopenhagenDenmark
| | - Andrea Malsam
- Heidelberg University Biochemistry CenterHeidelbergDenmark
| | - Jörg Malsam
- Heidelberg University Biochemistry CenterHeidelbergDenmark
| | - Fabio Lolicato
- Heidelberg University Biochemistry CenterHeidelbergDenmark
- Department of Physics, University of HelsinkiHelsinkiFinland
| | - Walter Nickel
- Heidelberg University Biochemistry CenterHeidelbergDenmark
| | | | | |
Collapse
|
7
|
Wu C, Ruan T, Yuan Y, Xu C, Du L, Wang F, Xu S. Alterations in Synaptic Connectivity and Synaptic Transmission in Alzheimer's Disease with High Physical Activity. J Alzheimers Dis 2024; 99:1005-1022. [PMID: 38759013 DOI: 10.3233/jad-240123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2024]
Abstract
Background Alzheimer's disease (AD) is a progressive neurodegeneration disease. Physical activity is one of the most promising modifiable lifestyles that can be effective in slowing down the progression of AD at an early stage. Objective Explore the molecular processes impaired in AD that were conversely preserved and enhanced by physical activity. Methods Integrated transcriptomic analyses were performed in datasets that contain AD patients and elders with different degrees of physical activity. The changes of the hub genes were validated through analyzing another two datasets. The expression of the hub genes was further detected in the hippocampus and cortexes of APP/PS1 transgenic mice with or without physical activity by Quantitative polymerase chain reaction (qPCR). Results Cross-comparison highlighted 195 DEGs displaying opposed regulation patterns between AD and high physical activity (HPA). The common DEGs were predominantly involved in synaptic vesicle recycling and synaptic transmission, largely downregulated in AD patients but upregulated in the elders with HPA. Two key modules and four hub genes that were related to synaptic vesicle turnover were obtained from the PPI network. The expression of these hub genes (SYT1, SYT4, SH3GL2, and AP2M1) was significantly decreased in AD transgenic mice and was reversed by HPA training. Conclusions HPA may reverse AD pathology by upregulating a range of synaptic vesicle transport related proteins which might improve the efficiency of synaptic vesicle turnover and facilitate inter-neuronal information transfer. The study provides novel insights into the mechanisms underlining the protective effects of HPA on AD.
Collapse
Affiliation(s)
- Can Wu
- Department of Physiology and Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Tingting Ruan
- Department of Physiology and Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Yalan Yuan
- Department of Physiology and Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Chunshuang Xu
- Department of Physiology and Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Lijuan Du
- Department of Physiology and Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- Faculty of Physical Education, Ningbo University, Ningbo, Zhejiang, China
| | - Fang Wang
- Department of Pharmacy, Zhejiang Pharmaceutical University, Ningbo, Zhejiang, China
| | - Shujun Xu
- Department of Physiology and Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
8
|
Vasu SO, Kaphzan H. Direct Current Stimulation Modulates Synaptic Facilitation via Distinct Presynaptic Calcium Channels. Int J Mol Sci 2023; 24:16866. [PMID: 38069188 PMCID: PMC10706473 DOI: 10.3390/ijms242316866] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/23/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Transcranial direct current stimulation (tDCS) is a subthreshold neurostimulation technique known for ameliorating neuropsychiatric conditions. The principal mechanism of tDCS is the differential polarization of subcellular neuronal compartments, particularly the axon terminals that are sensitive to external electrical fields. Yet, the underlying mechanism of tDCS is not fully clear. Here, we hypothesized that direct current stimulation (DCS)-induced modulation of presynaptic calcium channel conductance alters axon terminal dynamics with regard to synaptic vesicle release. To examine the involvement of calcium-channel subtypes in tDCS, we recorded spontaneous excitatory postsynaptic currents (sEPSCs) from cortical layer-V pyramidal neurons under DCS while selectively inhibiting distinct subtypes of voltage-dependent calcium channels. Blocking P/Q or N-type calcium channels occluded the effects of DCS on sEPSCs, demonstrating their critical role in the process of DCS-induced modulation of spontaneous vesicle release. However, inhibiting T-type calcium channels did not occlude DCS-induced modulation of sEPSCs, suggesting that despite being active in the subthreshold range, T-type calcium channels are not involved in the axonal effects of DCS. DCS modulates synaptic facilitation by regulating calcium channels in axon terminals, primarily via controlling P/Q and N-type calcium channels, while T-type calcium channels are not involved in this mechanism.
Collapse
Affiliation(s)
| | - Hanoch Kaphzan
- Sagol Department of Neurobiology, University of Haifa, Haifa 3103301, Israel
| |
Collapse
|
9
|
Wang CS, Monteggia LM, Kavalali ET. Spatially non-overlapping Ca 2+ signals drive distinct forms of neurotransmission. Cell Rep 2023; 42:113201. [PMID: 37777959 PMCID: PMC10842353 DOI: 10.1016/j.celrep.2023.113201] [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: 03/08/2023] [Revised: 06/23/2023] [Accepted: 09/16/2023] [Indexed: 10/03/2023] Open
Abstract
Calcium (Ca2+) signaling is tightly regulated within a presynaptic bouton. Here, we visualize Ca2+ signals within hippocampal presynaptic boutons using GCaMP8s tagged to synaptobrevin, a synaptic vesicle protein. We identify evoked presynaptic Ca2+ transients (ePreCTs) that derive from synchronized voltage-gated Ca2+ channel openings, spontaneous presynaptic Ca2+ transients (sPreCTs) that originate from ryanodine sensitive Ca2+ stores, and a baseline Ca2+ signal that arises from stochastic voltage-gated Ca2+ channel openings. We find that baseline Ca2+, but not sPreCTs, contributes to spontaneous glutamate release. We employ photobleaching as a use-dependent tool to probe nano-organization of Ca2+ signals and observe that all three occur in non-overlapping domains within the synapse at near-resting conditions. However, increased depolarization induces intermixing of these Ca2+ domains via both local and non-local synaptic vesicle turnover. Our findings reveal nanosegregation of Ca2+ signals within a presynaptic terminal that derive from multiple sources and in turn drive specific modes of neurotransmission.
Collapse
Affiliation(s)
- Camille S Wang
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 3729-7933, USA
| | - Lisa M Monteggia
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 3729-7933, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA
| | - Ege T Kavalali
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 3729-7933, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA.
| |
Collapse
|
10
|
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
|
11
|
Machamer JB, Vazquez-Cintron EJ, Stenslik MJ, Pagarigan KT, Bradford AB, Ondeck CA, McNutt PM. Neuromuscular recovery from botulism involves multiple forms of compensatory plasticity. Front Cell Neurosci 2023; 17:1226194. [PMID: 37650071 PMCID: PMC10463753 DOI: 10.3389/fncel.2023.1226194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 07/28/2023] [Indexed: 09/01/2023] Open
Abstract
Introduction Botulinum neurotoxin (BoNT) causes neuroparalytic disease and death by blocking neuromuscular transmission. There are no specific therapies for clinical botulism and the only treatment option is supportive care until neuromuscular function spontaneously recovers, which can take weeks or months after exposure. The highly specialized neuromuscular junction (NMJ) between phrenic motor neurons and diaphragm muscle fibers is the main clinical target of BoNT. Due to the difficulty in eliciting respiratory paralysis without a high mortality rate, few studies have characterized the neurophysiological mechanisms involved in diaphragm recovery from intoxication. Here, we develop a mouse model of botulism that involves partial paralysis of respiratory muscles with low mortality rates, allowing for longitudinal analysis of recovery. Methods and results Mice challenged by systemic administration of 0.7 LD50 BoNT/A developed physiological signs of botulism, such as respiratory depression and reduced voluntary running activity, that persisted for an average of 8-12 d. Studies in isolated hemidiaphragm preparations from intoxicated mice revealed profound reductions in nerve-elicited, tetanic and twitch muscle contraction strengths that recovered to baseline 21 d after intoxication. Despite apparent functional recovery, neurophysiological parameters remained depressed for 28 d, including end plate potential (EPP) amplitude, EPP success rate, quantal content (QC), and miniature EPP (mEPP) frequency. However, QC recovered more quickly than mEPP frequency, which could explain the discrepancy between muscle function studies and neurophysiological recordings. Hypothesizing that differential modulation of voltage-gated calcium channels (VGCC) contributed to the uncoupling of QC from mEPP frequency, pharmacological inhibition studies were used to study the contributions of different VGCCs to neurophysiological function. We found that N-type VGCC and P/Q-type VGCC partially restored QC but not mEPP frequency during recovery from paralysis, potentially explaining the accelerated recovery of evoked release versus spontaneous release. We identified additional changes that presumably compensate for reduced acetylcholine release during recovery, including increased depolarization of muscle fiber resting membrane potential and increased quantal size. Discussion In addition to identifying multiple forms of compensatory plasticity that occur in response to reduced NMJ function, it is expected that insights into the molecular mechanisms involved in recovery from neuromuscular paralysis will support new host-targeted treatments for multiple neuromuscular diseases.
Collapse
Affiliation(s)
- James B. Machamer
- BASF, Research Triangle Park, NC, United States
- United States Army Medical Research Institute of Chemical Defense, Gunpowder, MD, United States
| | | | - Mallory J. Stenslik
- United States Army Medical Research Institute of Chemical Defense, Gunpowder, MD, United States
| | - Kathleen T. Pagarigan
- United States Army Medical Research Institute of Chemical Defense, Gunpowder, MD, United States
| | - Aaron B. Bradford
- United States Army Medical Research Institute of Chemical Defense, Gunpowder, MD, United States
| | - Celinia A. Ondeck
- United States Army Medical Research Institute of Chemical Defense, Gunpowder, MD, United States
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Patrick M. McNutt
- United States Army Medical Research Institute of Chemical Defense, Gunpowder, MD, United States
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
12
|
Rahman MM, Pathak A, Schueler KL, Alsharif H, Michl A, Alexander J, Kim JA, Bhatnagar S. Genetic ablation of synaptotagmin-9 alters tomosyn-1 function to increase insulin secretion from pancreatic β-cells improving glucose clearance. FASEB J 2023; 37:e23075. [PMID: 37432648 PMCID: PMC10348599 DOI: 10.1096/fj.202300291rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/12/2023]
Abstract
Stimulus-coupled insulin secretion from the pancreatic islet β-cells involves the fusion of insulin granules to the plasma membrane (PM) via SNARE complex formation-a cellular process key for maintaining whole-body glucose homeostasis. Less is known about the role of endogenous inhibitors of SNARE complexes in insulin secretion. We show that an insulin granule protein synaptotagmin-9 (Syt9) deletion in mice increased glucose clearance and plasma insulin levels without affecting insulin action compared to the control mice. Upon glucose stimulation, increased biphasic and static insulin secretion were observed from ex vivo islets due to Syt9 loss. Syt9 colocalizes and binds with tomosyn-1 and the PM syntaxin-1A (Stx1A); Stx1A is required for forming SNARE complexes. Syt9 knockdown reduced tomosyn-1 protein abundance via proteasomal degradation and binding of tomosyn-1 to Stx1A. Furthermore, Stx1A-SNARE complex formation was increased, implicating Syt9-tomosyn-1-Stx1A complex is inhibitory in insulin secretion. Rescuing tomosyn-1 blocked the Syt9-knockdown-mediated increases in insulin secretion. This shows that the inhibitory effects of Syt9 on insulin secretion are mediated by tomosyn-1. We report a molecular mechanism by which β-cells modulate their secretory capacity rendering insulin granules nonfusogenic by forming the Syt9-tomosyn-1-Stx1A complex. Altogether, Syt9 loss in β-cells decreases tomosyn-1 protein abundance, increasing the formation of Stx1A-SNARE complexes, insulin secretion, and glucose clearance. These outcomes differ from the previously published work that identified Syt9 has either a positive or no effect of Syt9 on insulin secretion. Future work using β-cell-specific deletion of Syt9 mice is key for establishing the role of Syt9 in insulin secretion.
Collapse
Affiliation(s)
- Md Mostafizur Rahman
- Heersink School of Medicine, Division of Endocrinology, Diabetes, & Metabolism, Comprehensive Diabetes Center, University of Alabama, Birmingham, AL, 35294
| | - Asmita Pathak
- Heersink School of Medicine, Division of Endocrinology, Diabetes, & Metabolism, Comprehensive Diabetes Center, University of Alabama, Birmingham, AL, 35294
| | | | - Haifa Alsharif
- Heersink School of Medicine, Division of Endocrinology, Diabetes, & Metabolism, Comprehensive Diabetes Center, University of Alabama, Birmingham, AL, 35294
| | - Ava Michl
- Heersink School of Medicine, Division of Endocrinology, Diabetes, & Metabolism, Comprehensive Diabetes Center, University of Alabama, Birmingham, AL, 35294
| | - Justin Alexander
- Heersink School of Medicine, Division of Endocrinology, Diabetes, & Metabolism, Comprehensive Diabetes Center, University of Alabama, Birmingham, AL, 35294
| | - Jeong-A Kim
- Heersink School of Medicine, Division of Endocrinology, Diabetes, & Metabolism, Comprehensive Diabetes Center, University of Alabama, Birmingham, AL, 35294
| | - Sushant Bhatnagar
- Heersink School of Medicine, Division of Endocrinology, Diabetes, & Metabolism, Comprehensive Diabetes Center, University of Alabama, Birmingham, AL, 35294
| |
Collapse
|
13
|
Beccano-Kelly DA, Cherubini M, Mousba Y, Cramb KM, Giussani S, Caiazza MC, Rai P, Vingill S, Bengoa-Vergniory N, Ng B, Corda G, Banerjee A, Vowles J, Cowley S, Wade-Martins R. Calcium dysregulation combined with mitochondrial failure and electrophysiological maturity converge in Parkinson's iPSC-dopamine neurons. iScience 2023; 26:107044. [PMID: 37426342 PMCID: PMC10329047 DOI: 10.1016/j.isci.2023.107044] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/30/2022] [Accepted: 06/01/2023] [Indexed: 07/11/2023] Open
Abstract
Parkinson's disease (PD) is characterized by a progressive deterioration of motor and cognitive functions. Although death of dopamine neurons is the hallmark pathology of PD, this is a late-stage disease process preceded by neuronal dysfunction. Here we describe early physiological perturbations in patient-derived induced pluripotent stem cell (iPSC)-dopamine neurons carrying the GBA-N370S mutation, a strong genetic risk factor for PD. GBA-N370S iPSC-dopamine neurons show an early and persistent calcium dysregulation notably at the mitochondria, followed by reduced mitochondrial membrane potential and oxygen consumption rate, indicating mitochondrial failure. With increased neuronal maturity, we observed decreased synaptic function in PD iPSC-dopamine neurons, consistent with the requirement for ATP and calcium to support the increase in electrophysiological activity over time. Our work demonstrates that calcium dyshomeostasis and mitochondrial failure impair the higher electrophysiological activity of mature neurons and may underlie the vulnerability of dopamine neurons in PD.
Collapse
Affiliation(s)
- Dayne A. Beccano-Kelly
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Marta Cherubini
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Yassine Mousba
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Kaitlyn M.L. Cramb
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Stefania Giussani
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Maria Claudia Caiazza
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Pavandeep Rai
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Siv Vingill
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Nora Bengoa-Vergniory
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Bryan Ng
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Gabriele Corda
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
| | - Abhirup Banerjee
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford OX3 9DU, UK
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford OX3 7DQ, UK
| | - Jane Vowles
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- The James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Sally Cowley
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- The James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Richard Wade-Martins
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX3 7BN, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| |
Collapse
|
14
|
Wender M, Bornschein G, Brachtendorf S, Hallermann S, Eilers J, Schmidt H. Ca v2.2 Channels Sustain Vesicle Recruitment at a Mature Glutamatergic Synapse. J Neurosci 2023; 43:4005-4018. [PMID: 37185239 PMCID: PMC10255130 DOI: 10.1523/jneurosci.1279-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 04/06/2023] [Accepted: 04/08/2023] [Indexed: 05/17/2023] Open
Abstract
The composition of voltage-gated Ca2+ channel (Cav) subtypes that gate action potential (AP)-evoked release changes during the development of mammalian CNS synapses. Cav2.2 and Cav2.3 lose their function in gating-evoked release during postnatal synapse maturation. In mature boutons, Cav2.1 currents provide the almost exclusive trigger for evoked release, and Cav2.3 currents are required for the induction of presynaptic long-term potentiation. However, the functional significance of Cav2.2 remained elusive in mature boutons, although they remain present at active zones and continue contributing significantly to presynaptic Ca2+ influx. Here, we addressed the functional significance of Cav2.2 and Cav2.3 at mature parallel-fiber (PF) to Purkinje neuron synapses of mice of either sex. These synapses are known to exhibit the corresponding developmental Cav subtype changes in gating release. We addressed two hypotheses, namely that Cav2.2 and Cav2.3 are involved in triggering spontaneous glutamate release and that they are engaged in vesicle recruitment during repetitive evoked release. We found that spontaneous miniature release is Ca2+ dependent. However, experiments with Cav subtype-specific blockers excluded the spontaneous opening of Cavs as the Ca2+ source for spontaneous glutamate release. Thus, neither Cav2.2 nor Cav2.3 controls spontaneous release from PF boutons. Furthermore, vesicle recruitment during brief bursts of APs was also independent of Ca2+ influx through Cav2.2 and Cav2.3. However, Cav2.2, but not Cav2.3, currents significantly boosted vesicle recruitment during sustained high-frequency synaptic transmission. Thus, in mature PF boutons Cav2.2 channels are specifically required to sustain synaptic transmission during prolonged neuronal activity.SIGNIFICANCE STATEMENT At young CNS synapses, action potential-evoked release is gated via three subtypes of voltage-gated Ca2+ channels: Cav2.1, Cav2.2, and Cav2.3. During postnatal maturation, Cav2.2 and Cav2.3 lose their function in gating evoked release, such that at mature synapses Cav2.1 provides the almost exclusive source for triggering evoked release. Cav2.3 currents are required for the induction of presynaptic long-term potentiation. However, the function of the still abundant Cav2.2 in mature boutons remained largely elusive. Here, we studied mature cerebellar parallel-fiber synapses and found that Cav2.2 does not control spontaneous release. However, Ca2+ influx through Cav2.2 significantly boosted vesicle recruitment during trains of action potentials. Thus, Cav2.2 in mature parallel-fiber boutons participate in sustaining synaptic transmission during prolonged activity.
Collapse
Affiliation(s)
- Magdalena Wender
- Carl Ludwig Institute for Physiology, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Grit Bornschein
- Carl Ludwig Institute for Physiology, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Simone Brachtendorf
- Carl Ludwig Institute for Physiology, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Stefan Hallermann
- Carl Ludwig Institute for Physiology, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Jens Eilers
- Carl Ludwig Institute for Physiology, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Hartmut Schmidt
- Carl Ludwig Institute for Physiology, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
15
|
Chang HF, Schirra C, Pattu V, Krause E, Becherer U. Lytic granule exocytosis at immune synapses: lessons from neuronal synapses. Front Immunol 2023; 14:1177670. [PMID: 37275872 PMCID: PMC10233144 DOI: 10.3389/fimmu.2023.1177670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/09/2023] [Indexed: 06/07/2023] Open
Abstract
Regulated exocytosis is a central mechanism of cellular communication. It is not only the basis for neurotransmission and hormone release, but also plays an important role in the immune system for the release of cytokines and cytotoxic molecules. In cytotoxic T lymphocytes (CTLs), the formation of the immunological synapse is required for the delivery of the cytotoxic substances such as granzymes and perforin, which are stored in lytic granules and released via exocytosis. The molecular mechanisms of their fusion with the plasma membrane are only partially understood. In this review, we discuss the molecular players involved in the regulated exocytosis of CTL, highlighting the parallels and differences to neuronal synaptic transmission. Additionally, we examine the strengths and weaknesses of both systems to study exocytosis.
Collapse
|
16
|
Mackay JP, Smith-Dijak AI, Koch ET, Zhang P, Fung E, Nassrallah WB, Buren C, Schmidt M, Hayden MR, Raymond LA. Axonal ER Ca 2+ Release Selectively Enhances Activity-Independent Glutamate Release in a Huntington Disease Model. J Neurosci 2023; 43:3743-3763. [PMID: 36944490 PMCID: PMC10198457 DOI: 10.1523/jneurosci.1593-22.2023] [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: 08/19/2022] [Revised: 03/08/2023] [Accepted: 03/15/2023] [Indexed: 03/23/2023] Open
Abstract
Action potential (AP)-independent (miniature) neurotransmission occurs at all chemical synapses but remains poorly understood, particularly in pathologic contexts. Axonal endoplasmic reticulum (ER) Ca2+ stores are thought to influence miniature neurotransmission, and aberrant ER Ca2+ handling is implicated in progression of Huntington disease (HD). Here, we report elevated mEPSC frequencies in recordings from YAC128 mouse (HD-model) neurons (from cortical cultures and striatum-containing brain slices, both from male and female animals). Pharmacological experiments suggest that this is mediated indirectly by enhanced tonic ER Ca2+ release. Calcium imaging, using an axon-localized sensor, revealed slow AP-independent ER Ca2+ release waves in both YAC128 and WT cultures. These Ca2+ waves occurred at similar frequencies in both genotypes but spread less extensively and were of lower amplitude in YAC128 axons, consistent with axonal ER Ca2+ store depletion. Surprisingly, basal cytosolic Ca2+ levels were lower in YAC128 boutons and YAC128 mEPSCs were less sensitive to intracellular Ca2+ chelation. Together, these data suggest that elevated miniature glutamate release in YAC128 cultures is associated with axonal ER Ca2+ depletion but not directly mediated by ER Ca2+ release into the cytoplasm. In contrast to increased mEPSC frequencies, cultured YAC128 cortical neurons showed less frequent AP-dependent (spontaneous) Ca2+ events in soma and axons, although evoked glutamate release detected by an intensity-based glutamate-sensing fluorescence reporter in brain slices was similar between genotypes. Our results indicate that axonal ER dysfunction selectively elevates miniature glutamate release from cortical terminals in HD. This, together with reduced spontaneous cortical neuron firing, may cause a shift from activity-dependent to -independent glutamate release in HD, with potential implications for fidelity and plasticity of cortical excitatory signaling.SIGNIFICANCE STATEMENT Miniature neurotransmitter release persists at all chemical neuronal synapses in the absence of action potential firing but remains poorly understood, particularly in disease states. We show enhanced miniature glutamate release from cortical neurons in the YAC128 mouse Huntington disease model. This effect is mediated by axonal ER Ca2+ store depletion, but is not obviously due to elevated ER-to-cytosol Ca2+ release. Conversely, YAC128 cortical pyramidal neurons fired fewer action potentials and evoked cortical glutamate release was similar between WT an YAC128 preparations, indicating axonal ER depletion selectively enhances miniature glutamate release in YAC128 mice. These results extend our understanding of action potential independent neurotransmission and highlight a potential involvement of elevated miniature glutamate release in Huntington disease pathology.
Collapse
Affiliation(s)
- James P Mackay
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health
| | - Amy I Smith-Dijak
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health
- Graduate Program in Neuroscience
| | - Ellen T Koch
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health
- Graduate Program in Neuroscience
| | - Peng Zhang
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Evan Fung
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health
| | - Wissam B Nassrallah
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health
- MD/PhD Program
| | - Caodu Buren
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health
- Graduate Program in Neuroscience
| | - Mandi Schmidt
- Graduate Program in Neuroscience
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Michael R Hayden
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Lynn A Raymond
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health
| |
Collapse
|
17
|
Yang XK, Zhang FL, Jin XK, Jiao YT, Zhang XW, Liu YL, Amatore C, Huang WH. Nanoelectrochemistry reveals how soluble Aβ 42 oligomers alter vesicular storage and release of glutamate. Proc Natl Acad Sci U S A 2023; 120:e2219994120. [PMID: 37126689 PMCID: PMC10175745 DOI: 10.1073/pnas.2219994120] [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: 11/23/2022] [Accepted: 03/30/2023] [Indexed: 05/03/2023] Open
Abstract
Glutamate (Glu) is the major excitatory transmitter in the nervous system. Impairment of its vesicular release by β-amyloid (Aβ) oligomers is thought to participate in pathological processes leading to Alzheimer's disease. However, it remains unclear whether soluble Aβ42 oligomers affect intravesicular amounts of Glu or their release in the brain, or both. Measurements made in this work on single Glu varicosities with an amperometric nanowire Glu biosensor revealed that soluble Aβ42 oligomers first caused a dramatic increase in vesicular Glu storage and stimulation-induced release, accompanied by a high level of parallel spontaneous exocytosis, ultimately resulting in the depletion of intravesicular Glu content and greatly reduced release. Molecular biology tools and mouse models of Aβ amyloidosis have further established that the transient hyperexcitation observed during the primary pathological stage is mediated by an altered behavior of VGLUT1 responsible for transporting Glu into synaptic vesicles. Thereafter, an overexpression of Vps10p-tail-interactor-1a, a protein that maintains spontaneous release of neurotransmitters by selective interaction with t-SNAREs, resulted in a depletion of intravesicular Glu content, triggering advanced-stage neuronal malfunction. These findings are expected to open perspectives for remediating Aβ42-induced neuronal hyperactivity and neuronal degeneration.
Collapse
Affiliation(s)
- Xiao-Ke Yang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan430072, People’s Republic of China
| | - Fu-Li Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan430072, People’s Republic of China
| | - Xue-Ke Jin
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan430072, People’s Republic of China
| | - Yu-Ting Jiao
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan430072, People’s Republic of China
| | - Xin-Wei Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan430072, People’s Republic of China
| | - Yan-Ling Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan430072, People’s Republic of China
| | - Christian Amatore
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen361005, People’s Republic of China
- PASTEUR, Département de Chimie, École Normale Supérieure, Paris Sciences Lettre Research University, Sorbonne University, & University Pierre and Marie Curie, 0675005Paris, France
| | - Wei-Hua Huang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan430072, People’s Republic of China
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan430071, People’s Republic of China
| |
Collapse
|
18
|
Grasskamp AT, Jusyte M, McCarthy AW, Götz TWB, Ditlevsen S, Walter AM. Spontaneous neurotransmission at evocable synapses predicts their responsiveness to action potentials. Front Cell Neurosci 2023; 17:1129417. [PMID: 36970416 PMCID: PMC10030884 DOI: 10.3389/fncel.2023.1129417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/16/2023] [Indexed: 03/29/2023] Open
Abstract
Synaptic transmission relies on presynaptic neurotransmitter (NT) release from synaptic vesicles (SVs) and on NT detection by postsynaptic receptors. Transmission exists in two principal modes: action-potential (AP) evoked and AP-independent, "spontaneous" transmission. AP-evoked neurotransmission is considered the primary mode of inter-neuronal communication, whereas spontaneous transmission is required for neuronal development, homeostasis, and plasticity. While some synapses appear dedicated to spontaneous transmission only, all AP-responsive synapses also engage spontaneously, but whether this encodes functional information regarding their excitability is unknown. Here we report on functional interdependence of both transmission modes at individual synaptic contacts of Drosophila larval neuromuscular junctions (NMJs) which were identified by the presynaptic scaffolding protein Bruchpilot (BRP) and whose activities were quantified using the genetically encoded Ca2+ indicator GCaMP. Consistent with the role of BRP in organizing the AP-dependent release machinery (voltage-dependent Ca2+ channels and SV fusion machinery), most active BRP-positive synapses (>85%) responded to APs. At these synapses, the level of spontaneous activity was a predictor for their responsiveness to AP-stimulation. AP-stimulation resulted in cross-depletion of spontaneous activity and both transmission modes were affected by the non-specific Ca2+ channel blocker cadmium and engaged overlapping postsynaptic receptors. Thus, by using overlapping machinery, spontaneous transmission is a continuous, stimulus independent predictor for the AP-responsiveness of individual synapses.
Collapse
Affiliation(s)
| | - Meida Jusyte
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
- Einstein Center for Neurosciences, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | | | - Torsten W. B. Götz
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Susanne Ditlevsen
- Department of Mathematical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alexander M. Walter
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
- Einstein Center for Neurosciences, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
19
|
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: 0] [Impact Index Per Article: 0] [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
|
20
|
López-Murcia FJ, Reim K, Taschenberger H. Complexins: Ubiquitously Expressed Presynaptic Regulators of SNARE-Mediated Synaptic Vesicle Fusion. ADVANCES IN NEUROBIOLOGY 2023; 33:255-285. [PMID: 37615870 DOI: 10.1007/978-3-031-34229-5_10] [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
Neurotransmitter release is a spatially and temporally tightly regulated process, which requires assembly and disassembly of SNARE complexes to enable the exocytosis of transmitter-loaded synaptic vesicles (SVs) at presynaptic active zones (AZs). While the requirement for the core SNARE machinery is shared by most membrane fusion processes, SNARE-mediated fusion at AZs is uniquely regulated to allow very rapid Ca2+-triggered SV exocytosis following action potential (AP) arrival. To enable a sub-millisecond time course of AP-triggered SV fusion, synapse-specific accessory SNARE-binding proteins are required in addition to the core fusion machinery. Among the known SNARE regulators specific for Ca2+-triggered SV fusion are complexins, which are almost ubiquitously expressed in neurons. This chapter summarizes the structural features of complexins, models for their molecular interactions with SNAREs, and their roles in SV fusion.
Collapse
Affiliation(s)
- Francisco José López-Murcia
- Department of Pathology and Experimental Therapy, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.
| | - Kerstin Reim
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| | - Holger Taschenberger
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| |
Collapse
|
21
|
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
|
22
|
Yeo XY, Lim YT, Chae WR, Park C, Park H, Jung S. Alterations of presynaptic proteins in autism spectrum disorder. Front Mol Neurosci 2022; 15:1062878. [DOI: 10.3389/fnmol.2022.1062878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 10/31/2022] [Indexed: 11/19/2022] Open
Abstract
The expanded use of hypothesis-free gene analysis methods in autism research has significantly increased the number of genetic risk factors associated with the pathogenesis of autism. A further examination of the implicated genes directly revealed the involvement in processes pertinent to neuronal differentiation, development, and function, with a predominant contribution from the regulators of synaptic function. Despite the importance of presynaptic function in synaptic transmission, the regulation of neuronal network activity, and the final behavioral output, there is a relative lack of understanding of the presynaptic contribution to the pathology of autism. Here, we will review the close association among autism-related mutations, autism spectrum disorders (ASD) phenotypes, and the altered presynaptic protein functions through a systematic examination of the presynaptic risk genes relating to the critical stages of synaptogenesis and neurotransmission.
Collapse
|
23
|
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: 31] [Impact Index Per Article: 15.5] [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
|
24
|
Zhu J, McDargh ZA, Li F, Krishnakumar SS, Rothman JE, O’Shaughnessy B. Synaptotagmin rings as high-sensitivity regulators of synaptic vesicle docking and fusion. Proc Natl Acad Sci U S A 2022; 119:e2208337119. [PMID: 36103579 PMCID: PMC9499556 DOI: 10.1073/pnas.2208337119] [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: 05/19/2022] [Accepted: 07/05/2022] [Indexed: 11/18/2022] Open
Abstract
Synchronous release at neuronal synapses is accomplished by a machinery that senses calcium influx and fuses the synaptic vesicle and plasma membranes to release neurotransmitters. Previous studies suggested the calcium sensor synaptotagmin (Syt) is a facilitator of vesicle docking and both a facilitator and inhibitor of fusion. On phospholipid monolayers, the Syt C2AB domain spontaneously oligomerized into rings that are disassembled by Ca2+, suggesting Syt rings may clamp fusion as membrane-separating "washers" until Ca2+-mediated disassembly triggers fusion and release [J. Wang et al., Proc. Natl. Acad. Sci. U.S.A. 111, 13966-13971 (2014)].). Here, we combined mathematical modeling with experiment to measure the mechanical properties of Syt rings and to test this mechanism. Consistent with experimental results, the model quantitatively recapitulates observed Syt ring-induced dome and volcano shapes on phospholipid monolayers and predicts rings are stabilized by anionic phospholipid bilayers or bulk solution with ATP. The selected ring conformation is highly sensitive to membrane composition and bulk ATP levels, a property that may regulate vesicle docking and fusion in ATP-rich synaptic terminals. We find the Syt molecules hosted by a synaptic vesicle oligomerize into a halo, unbound from the vesicle, but in proximity to sufficiently phosphatidylinositol 4,5-bisphosphate (PIP2)-rich plasma membrane (PM) domains, the PM-bound trans Syt ring conformation is preferred. Thus, the Syt halo serves as landing gear for spatially directed docking at PIP2-rich sites that define the active zones of exocytotic release, positioning the Syt ring to clamp fusion and await calcium. Our results suggest the Syt ring is both a Ca2+-sensitive fusion clamp and a high-fidelity sensor for directed docking.
Collapse
Affiliation(s)
- Jie Zhu
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520
| | - Zachary A. McDargh
- Department of Chemical Engineering, Columbia University, New York, NY 10027
| | - Feng Li
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520
| | | | - James E. Rothman
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520
| | - Ben O’Shaughnessy
- Department of Chemical Engineering, Columbia University, New York, NY 10027
| |
Collapse
|
25
|
Atlas D. Revisiting the molecular basis of synaptic transmission. Prog Neurobiol 2022; 216:102312. [PMID: 35760141 DOI: 10.1016/j.pneurobio.2022.102312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/12/2022] [Accepted: 06/20/2022] [Indexed: 10/17/2022]
Abstract
Measurements of the time elapsed during synaptic transmission has shown that synaptic vesicle (SV) fusion lags behind Ca2+-influx by approximately 60 microseconds (µsec). The conventional model cannot explain this extreme rapidity of the release event. Synaptic transmission occurs at the active zone (AZ), which comprises of two pools of SV, non-releasable "tethered" vesicles, and a readily-releasable pool of channel-associated Ca2+-primed vesicles, "RRP". A recent TIRF study at cerebellar-mossy fiber-terminal, showed that subsequent to an action potential, newly "tethered" vesicles, became fusion-competent in a Ca2+-dependent manner, 300-400 milliseconds after tethering, but were not fused. This time resolution may correspond to priming of tethered vesicles through Ca2+-binding to Syt1/Munc13-1/complexin. It confirms that Ca2+-priming and Ca2+-influx-independent fusion, are two distinct events. Notably, we have established that Ca2+ channel signals evoked-release in an ion flux-independent manner, demonstrated by Ca2+-impermeable channel, or a Ca2+ channel in which Ca2+ is replaced by impermeable La3+. Thus, conformational changes in a channel coupled to RRP appear to directly activate the release machinery and account for a µsec Ca2+-influx-independent vesicle fusion. Rapid vesicle fusion driven by non-ionotropic channel signaling strengthens a conformational-coupling mechanism of synaptic transmission, and contributes to better understanding of neuronal communication vital for brain function.
Collapse
Affiliation(s)
- Daphne Atlas
- Dept. of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, 91904 Israel.
| |
Collapse
|
26
|
Vardar G, Salazar-Lázaro A, Zobel S, Trimbuch T, Rosenmund C. Syntaxin-1A modulates vesicle fusion in mammalian neurons via juxtamembrane domain dependent palmitoylation of its transmembrane domain. eLife 2022; 11:78182. [PMID: 35638903 PMCID: PMC9183232 DOI: 10.7554/elife.78182] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/30/2022] [Indexed: 11/16/2022] Open
Abstract
SNAREs are undoubtedly one of the core elements of synaptic transmission. Contrary to the well characterized function of their SNARE domains bringing the plasma and vesicular membranes together, the level of contribution of their juxtamembrane domain (JMD) and the transmembrane domain (TMD) to the vesicle fusion is still under debate. To elucidate this issue, we analyzed three groups of STX1A mutations in cultured mouse hippocampal neurons: (1) elongation of STX1A’s JMD by three amino acid insertions in the junction of SNARE-JMD or JMD-TMD; (2) charge reversal mutations in STX1A’s JMD; and (3) palmitoylation deficiency mutations in STX1A’s TMD. We found that both JMD elongations and charge reversal mutations have position-dependent differential effects on Ca2+-evoked and spontaneous neurotransmitter release. Importantly, we show that STX1A’s JMD regulates the palmitoylation of STX1A’s TMD and loss of STX1A palmitoylation either through charge reversal mutation K260E or by loss of TMD cysteines inhibits spontaneous vesicle fusion. Interestingly, the retinal ribbon specific STX3B has a glutamate in the position corresponding to the K260E mutation in STX1A and mutating it with E259K acts as a molecular on-switch. Furthermore, palmitoylation of post-synaptic STX3A can be induced by the exchange of its JMD with STX1A’s JMD together with the incorporation of two cysteines into its TMD. Forced palmitoylation of STX3A dramatically enhances spontaneous vesicle fusion suggesting that STX1A regulates spontaneous release through two distinct mechanisms: one through the C-terminal half of its SNARE domain and the other through the palmitoylation of its TMD.
Collapse
Affiliation(s)
- Gülçin Vardar
- Department of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andrea Salazar-Lázaro
- Department of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sina Zobel
- Department of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Thorsten Trimbuch
- Department of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Rosenmund
- Department of Neurophysiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
27
|
Wang CS, Chanaday NL, Monteggia LM, Kavalali ET. Probing the segregation of evoked and spontaneous neurotransmission via photobleaching and recovery of a fluorescent glutamate sensor. eLife 2022; 11:e76008. [PMID: 35420542 PMCID: PMC9129874 DOI: 10.7554/elife.76008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 04/08/2022] [Indexed: 11/13/2022] Open
Abstract
Synapses maintain both action potential-evoked and spontaneous neurotransmitter release; however, organization of these two forms of release within an individual synapse remains unclear. Here, we used photobleaching properties of iGluSnFR, a fluorescent probe that detects glutamate, to investigate the subsynaptic organization of evoked and spontaneous release in primary hippocampal cultures. In nonneuronal cells and neuronal dendrites, iGluSnFR fluorescence is intensely photobleached and recovers via diffusion of nonphotobleached probes with a time constant of ~10 s. After photobleaching, while evoked iGluSnFR events could be rapidly suppressed, their recovery required several hours. In contrast, iGluSnFR responses to spontaneous release were comparatively resilient to photobleaching, unless the complete pool of iGluSnFR was activated by glutamate perfusion. This differential effect of photobleaching on different modes of neurotransmission is consistent with a subsynaptic organization where sites of evoked glutamate release are clustered and corresponding iGluSnFR probes are diffusion restricted, while spontaneous release sites are broadly spread across a synapse with readily diffusible iGluSnFR probes.
Collapse
Affiliation(s)
- Camille S Wang
- Vanderbilt Brain Institute, Vanderbilt UniversityNashvilleUnited States
| | - Natali L Chanaday
- Department of Pharmacology, Vanderbilt UniversityNashvilleUnited States
| | - Lisa M Monteggia
- Vanderbilt Brain Institute, Vanderbilt UniversityNashvilleUnited States
- Department of Pharmacology, Vanderbilt UniversityNashvilleUnited States
| | - Ege T Kavalali
- Vanderbilt Brain Institute, Vanderbilt UniversityNashvilleUnited States
- Department of Pharmacology, Vanderbilt UniversityNashvilleUnited States
| |
Collapse
|
28
|
Melland H, Bumbak F, Kolesnik-Taylor A, Ng-Cordell E, John A, Constantinou P, Joss S, Larsen M, Fagerberg C, Laulund LW, Thies J, Emslie F, Willemsen M, Kleefstra T, Pfundt R, Barrick R, Chang R, Loong L, Alfadhel M, van der Smagt J, Nizon M, Kurian MA, Scott DJ, Ziarek JJ, Gordon SL, Baker K. Expanding the genotype and phenotype spectrum of SYT1-associated neurodevelopmental disorder. Genet Med 2022; 24:880-893. [PMID: 35101335 PMCID: PMC8986325 DOI: 10.1016/j.gim.2021.12.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 12/03/2021] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Synaptotagmin-1 (SYT1) is a critical mediator of neurotransmitter release in the central nervous system. Previously reported missense SYT1 variants in the C2B domain are associated with severe intellectual disability, movement disorders, behavioral disturbances, and electroencephalogram abnormalities. In this study, we expand the genotypes and phenotypes and identify discriminating features of this disorder. METHODS We describe 22 individuals with 15 de novo missense SYT1 variants. The evidence for pathogenicity is discussed, including the American College of Medical Genetics and Genomics/Association for Molecular Pathology criteria, known structure-function relationships, and molecular dynamics simulations. Quantitative behavioral data for 14 cases were compared with other monogenic neurodevelopmental disorders. RESULTS Four variants were located in the C2A domain with the remainder in the C2B domain. We classified 6 variants as pathogenic, 4 as likely pathogenic, and 5 as variants of uncertain significance. Prevalent clinical phenotypes included delayed developmental milestones, abnormal eye physiology, movement disorders, and sleep disturbances. Discriminating behavioral characteristics were severity of motor and communication impairment, presence of motor stereotypies, and mood instability. CONCLUSION Neurodevelopmental disorder-associated SYT1 variants extend beyond previously reported regions, and the phenotypic spectrum encompasses a broader range of severities than initially reported. This study guides the diagnosis and molecular understanding of this rare neurodevelopmental disorder and highlights a key role for SYT1 function in emotional regulation, motor control, and emergent cognitive function.
Collapse
Affiliation(s)
- Holly Melland
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia; Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Fabian Bumbak
- Department of Molecular and Cellular Biochemistry, College of Arts + Sciences, Indiana University Bloomington, Bloomington, IN
| | - Anna Kolesnik-Taylor
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, United Kingdom
| | - Elise Ng-Cordell
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, United Kingdom
| | - Abinayah John
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, United Kingdom
| | - Panayiotis Constantinou
- Department of Clinical Genetics, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Shelagh Joss
- Department of Clinical Genetics, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Martin Larsen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Christina Fagerberg
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Lone Walentin Laulund
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark
| | - Jenny Thies
- Department of Pediatrics, Division of Genetic Medicine, Seattle Children's Hospital, Seattle, WA
| | - Frances Emslie
- South West Thames Regional Genetics Service and St George's University of London, London, United Kingdom
| | | | - Tjitske Kleefstra
- Radboud University Medical Center, Nijmegen, The Netherlands; Vincent van Gogh Centre for Neuropsychiatry, Venray, The Netherlands
| | - Rolf Pfundt
- Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | | - Lucy Loong
- Oxford Centre for Genomic Medicine, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Majid Alfadhel
- Genetics and Precision Medicine department, King Abdullah Specialized Children Hospital, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia; Medical Genomics Research Department, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | | | - Mathilde Nizon
- Service de Génétique Médicale, CHU de Nantes, INSERM, Université de Nantes, Nantes, France
| | - Manju A Kurian
- Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Daniel J Scott
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Joshua J Ziarek
- Department of Molecular and Cellular Biochemistry, College of Arts + Sciences, Indiana University Bloomington, Bloomington, IN
| | - Sarah L Gordon
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia; Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Kate Baker
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, United Kingdom; Department of Medical Genetics, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
29
|
Vasu SO, Kaphzan H. Calcium channels control tDCS-induced spontaneous vesicle release from axon terminals. Brain Stimul 2022; 15:270-282. [DOI: 10.1016/j.brs.2022.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/06/2022] [Accepted: 01/06/2022] [Indexed: 12/29/2022] Open
|
30
|
Patzke C, Dai J, Brockmann MM, Sun Z, Fenske P, Rosenmund C, Südhof TC. Cannabinoid receptor activation acutely increases synaptic vesicle numbers by activating synapsins in human synapses. Mol Psychiatry 2021; 26:6253-6268. [PMID: 33931733 DOI: 10.1038/s41380-021-01095-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 03/20/2021] [Accepted: 03/31/2021] [Indexed: 02/03/2023]
Abstract
Cannabis and cannabinoid drugs are central agents that are used widely recreationally and are employed broadly for treating psychiatric conditions. Cannabinoids primarily act by stimulating presynaptic CB1 receptors (CB1Rs), the most abundant G-protein-coupled receptors in brain. CB1R activation decreases neurotransmitter release by inhibiting presynaptic Ca2+ channels and induces long-term plasticity by decreasing cellular cAMP levels. Here we identified an unanticipated additional mechanism of acute cannabinoid signaling in presynaptic terminals that regulates the size of synaptic vesicle pools available for neurotransmitter release. Specifically, we show that activation of CB1Rs in human and mouse neurons rapidly recruits vesicles to nerve terminals by suppressing the cAMP-dependent phosphorylation of synapsins. We confirmed this unanticipated mechanism using conditional deletion of synapsin-1, the predominant synapsin isoform in human neurons, demonstrating that synapsin-1 significantly contributes to the CB1R-dependent regulation of neurotransmission. Interestingly, acute activation of the Gi-DREADD hM4D mimics the effect of CB1R activation in a synapsin-1-dependent manner, suggesting that the control of synaptic vesicle numbers by synapsin-1 phosphorylation is a general presynaptic mechanism of neuromodulation. Thus, we uncovered a CB1R-dependent presynaptic mechanism that rapidly regulates the organization and neurotransmitter release properties of synapses.
Collapse
Affiliation(s)
- Christopher Patzke
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA, 94305, USA. .,Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, 109A Galvin Life Science Center, Notre Dame, IN, 46556, USA.
| | - Jinye Dai
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA, 94305, USA.,Howard Hughes Medical Institute, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA, 94305, USA
| | - Marisa M Brockmann
- Institute of Neurophysiology, Charité Universitätsmedizin, 10117, Berlin, Germany
| | - Zijun Sun
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA, 94305, USA
| | - Pascal Fenske
- Institute of Neurophysiology, Charité Universitätsmedizin, 10117, Berlin, Germany
| | - Christian Rosenmund
- Institute of Neurophysiology, Charité Universitätsmedizin, 10117, Berlin, Germany
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA, 94305, USA.,Howard Hughes Medical Institute, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA, 94305, USA
| |
Collapse
|
31
|
Lee BJ, Yang CH, Lee SY, Lee SH, Kim Y, Ho WK. Voltage-gated calcium channels contribute to spontaneous glutamate release directly via nanodomain coupling or indirectly via calmodulin. Prog Neurobiol 2021; 208:102182. [PMID: 34695543 DOI: 10.1016/j.pneurobio.2021.102182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/05/2021] [Accepted: 10/18/2021] [Indexed: 11/19/2022]
Abstract
Neurotransmitter release occurs either synchronously with action potentials (evoked release) or spontaneously (spontaneous release). Whether the molecular mechanisms underlying evoked and spontaneous release are identical, especially whether voltage-gated calcium channels (VGCCs) can trigger spontaneous events, is still a matter of debate in glutamatergic synapses. To elucidate this issue, we characterized the VGCC dependence of miniature excitatory postsynaptic currents (mEPSCs) in various synapses with different coupling distances between VGCCs and synaptic vesicles, known as a critical factor in evoked release. We found that most of the extracellular calcium-dependent mEPSCs were attributable to VGCCs in cultured autaptic hippocampal neurons and the mature calyx of Held where VGCCs and vesicles were tightly coupled. Among loosely coupled synapses, mEPSCs were not VGCC-dependent at immature calyx of Held and CA1 pyramidal neuron synapses, whereas VGCCs contribution was significant at CA3 pyramidal neuron synapses. Interestingly, the contribution of VGCCs to spontaneous glutamate release in CA3 pyramidal neurons was abolished by a calmodulin antagonist, calmidazolium. These data suggest that coupling distance between VGCCs and vesicles determines VGCC dependence of spontaneous release at tightly coupled synapses, yet VGCC contribution can be achieved indirectly at loosely coupled synapses.
Collapse
Affiliation(s)
- Byoung Ju Lee
- Department of Biomedical Sciences, Seoul National University College of Natural Science, Seoul, Republic of Korea; Department of Physiology, Seoul National University College of Natural Science, Seoul, Republic of Korea
| | - Che Ho Yang
- Department of Biomedical Sciences, Seoul National University College of Natural Science, Seoul, Republic of Korea; Department of Physiology, Seoul National University College of Natural Science, Seoul, Republic of Korea; Department of Brain and Cognitive Science, Seoul National University College of Natural Science, Seoul, Republic of Korea
| | - Seung Yeon Lee
- Department of Biomedical Sciences, Seoul National University College of Natural Science, Seoul, Republic of Korea; Department of Physiology, Seoul National University College of Natural Science, Seoul, Republic of Korea
| | - Suk-Ho Lee
- Department of Biomedical Sciences, Seoul National University College of Natural Science, Seoul, Republic of Korea; Department of Physiology, Seoul National University College of Natural Science, Seoul, Republic of Korea; Neuroscience Research Institute, Seoul National University College of Medicine, Republic of Korea; Department of Brain and Cognitive Science, Seoul National University College of Natural Science, Seoul, Republic of Korea
| | - Yujin Kim
- Department of Physiology, Seoul National University College of Natural Science, Seoul, Republic of Korea; Neuroscience Research Institute, Seoul National University College of Medicine, Republic of Korea; Department of Brain and Cognitive Science, Seoul National University College of Natural Science, Seoul, Republic of Korea.
| | - Won-Kyung Ho
- Department of Biomedical Sciences, Seoul National University College of Natural Science, Seoul, Republic of Korea; Department of Physiology, Seoul National University College of Natural Science, Seoul, Republic of Korea; Neuroscience Research Institute, Seoul National University College of Medicine, Republic of Korea; Department of Brain and Cognitive Science, Seoul National University College of Natural Science, Seoul, Republic of Korea.
| |
Collapse
|
32
|
Li WR, Wang YL, Li C, Gao P, Zhang FF, Hu M, Li JC, Zhang S, Li R, Zhang CX. Synaptotagmin-11 inhibits spontaneous neurotransmission through vti1a. J Neurochem 2021; 159:729-741. [PMID: 34599505 DOI: 10.1111/jnc.15523] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 11/25/2020] [Accepted: 09/26/2021] [Indexed: 12/25/2022]
Abstract
Recent work has revealed that spontaneous release plays critical roles in the central nervous system, but how it is regulated remains elusive. Here, we report that synaptotagmin-11 (Syt11), a Ca2+ -independent Syt isoform associated with schizophrenia and Parkinson's disease, suppressed spontaneous release. Syt11-knockout hippocampal neurons showed an increased frequency of miniature excitatory post-synaptic currents while over-expression of Syt11 inversely decreased the frequency. Neither knockout nor over-expression of Syt11 affected the average amplitude, suggesting the pre-synaptic regulation of spontaneous neurotransmission by Syt11. Glutathione S-transferase pull-down, co-immunoprecipitation, and affinity-purification experiments demonstrated a direct interaction of Syt11 with vps10p-tail-interactor-1a (vti1a), a non-canonical SNARE protein that maintains spontaneous release. Importantly, knockdown of vti1a reversed the phenotype of Syt11 knockout, identifying vti1a as the main target of Syt11 inhibition. Domain analysis revealed that the C2A domain of Syt11 bound vti1a with high affinity. Consistently, expression of the C2A domain alone rescued the phenotype of elevated spontaneous release in Syt11-knockout neurons similar to the full-length protein. Altogether, our results suggest that Syt11 inhibits vti1a-containing vesicles during spontaneous release.
Collapse
Affiliation(s)
- Wan-Ru Li
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Ya-Long Wang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Chao Li
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Pei Gao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Fei-Fan Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Meiqin Hu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jing-Chen Li
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Shuli Zhang
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Rena Li
- Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital and Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Claire Xi Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.,Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| |
Collapse
|
33
|
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: 2.0] [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
|
34
|
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: 3.7] [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
|
35
|
Príncipe C, Dionísio de Sousa IJ, Prazeres H, Soares P, Lima RT. LRP1B: A Giant Lost in Cancer Translation. Pharmaceuticals (Basel) 2021; 14:836. [PMID: 34577535 PMCID: PMC8469001 DOI: 10.3390/ph14090836] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 08/14/2021] [Accepted: 08/16/2021] [Indexed: 12/23/2022] Open
Abstract
Low-density lipoprotein receptor-related protein 1B (LRP1B) is a giant member of the LDLR protein family, which includes several structurally homologous cell surface receptors with a wide range of biological functions from cargo transport to cell signaling. LRP1B is among the most altered genes in human cancer overall. Found frequently inactivated by several genetic and epigenetic mechanisms, it has mostly been regarded as a putative tumor suppressor. Still, limitations in LRP1B studies exist, in particular associated with its huge size. Therefore, LRP1B expression and function in cancer remains to be fully unveiled. This review addresses the current understanding of LRP1B and the studies that shed a light on the LRP1B structure and ligands. It goes further in presenting increasing knowledge brought by technical and methodological advances that allow to better manipulate LRP1B expression in cells and to more thoroughly explore its expression and mutation status. New evidence is pushing towards the increased relevance of LRP1B in cancer as a potential target or translational prognosis and response to therapy biomarker.
Collapse
Affiliation(s)
- Catarina Príncipe
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.P.); (H.P.); (P.S.)
- Cancer Signalling and Metabolism Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Isabel J. Dionísio de Sousa
- Department of Oncology, Centro Hospitalar Universitário de São João, 4200-450 Porto, Portugal;
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Hugo Prazeres
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.P.); (H.P.); (P.S.)
- IPO-Coimbra, Portuguese Oncology Institute of Coimbra, 3000-075 Coimbra, Portugal
| | - Paula Soares
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.P.); (H.P.); (P.S.)
- Cancer Signalling and Metabolism Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Raquel T. Lima
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.P.); (H.P.); (P.S.)
- Cancer Signalling and Metabolism Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
36
|
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: 1.3] [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
|
37
|
Polli FS, Roncacè V, Maximov A. The multifaceted role of SOCE in central synapses. Cell Calcium 2021; 97:102420. [PMID: 34022471 DOI: 10.1016/j.ceca.2021.102420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 05/11/2021] [Indexed: 11/27/2022]
Abstract
Store-operated calcium entry (SOCE) is a mechanism for calcium influx through the plasma membrane in response to release of free calcium from the endoplasmic reticulum. Two recent studies revealed how SOCE regulates the exocytosis of neurotransmitter vesicles at central synapses.
Collapse
Affiliation(s)
- Filip Souza Polli
- Department of Neuroscience and The Dorris Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Vincenzo Roncacè
- Department of Neuroscience and The Dorris Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Anton Maximov
- Department of Neuroscience and The Dorris Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
38
|
Chanaday NL, Nosyreva E, Shin OH, Zhang H, Aklan I, Atasoy D, Bezprozvanny I, Kavalali ET. Presynaptic store-operated Ca 2+ entry drives excitatory spontaneous neurotransmission and augments endoplasmic reticulum stress. Neuron 2021; 109:1314-1332.e5. [PMID: 33711258 PMCID: PMC8068669 DOI: 10.1016/j.neuron.2021.02.023] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 01/18/2021] [Accepted: 02/19/2021] [Indexed: 12/11/2022]
Abstract
Store-operated calcium entry (SOCE) is activated by depletion of Ca2+ from the endoplasmic reticulum (ER) and mediated by stromal interaction molecule (STIM) proteins. Here, we show that in rat and mouse hippocampal neurons, acute ER Ca2+ depletion increases presynaptic Ca2+ levels and glutamate release through a pathway dependent on STIM2 and the synaptic Ca2+ sensor synaptotagmin-7 (syt7). In contrast, synaptotagmin-1 (syt1) can suppress SOCE-mediated spontaneous release, and STIM2 is required for the increase in spontaneous release seen during syt1 loss of function. We also demonstrate that chronic ER stress activates the same pathway leading to syt7-dependent potentiation of spontaneous glutamate release. During ER stress, inhibition of SOCE or syt7-driven fusion partially restored basal neurotransmission and decreased expression of pro-apoptotic markers, indicating that these processes participate in the amplification of ER-stress-related damage. Taken together, we propose that presynaptic SOCE links ER stress and augmented spontaneous neurotransmission, which may, in turn, facilitate neurodegeneration.
Collapse
Affiliation(s)
- Natali L. Chanaday
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, 37240-7933, USA
| | - Elena Nosyreva
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Ok-Ho Shin
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, 37240-7933, USA
| | - Hua Zhang
- Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, TX 75390-9040, USA
| | - Iltan Aklan
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Deniz Atasoy
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA,Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA,FOE Diabetes Research Center, University of Iowa, Iowa City, IA, 52242, USA
| | - Ilya Bezprozvanny
- Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, TX 75390-9040, USA.,Laboratory of Molecular Neurodegeneration, Peter the Great St Petersburg State Polytechnic University, St. Petersburg, Russia
| | - Ege T. Kavalali
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, 37240-7933, USA.,Vanderbilt Brain Institute.,Corresponding author: Ege T. Kavalali, Ph.D., Department of Pharmacology, Vanderbilt University, 465 21st Avenue South, 7130A MRBIII, PMB407933 Nashville, TN 37240-7933, phone: 615-343-5480,
| |
Collapse
|
39
|
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: 1.7] [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
|
40
|
Dong X, Yang L, Liu K, Ji X, Tang C, Li W, Ma L, Mei Y, Peng T, Feng B, Wu Z, Tang Q, Gao Y, Yan K, Zhou W, Xiong M. Transcriptional networks identify synaptotagmin-like 3 as a regulator of cortical neuronal migration during early neurodevelopment. Cell Rep 2021; 34:108802. [PMID: 33657377 DOI: 10.1016/j.celrep.2021.108802] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 12/25/2020] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
Human brain development is a complex process involving neural proliferation, differentiation, and migration that are directed by many essential cellular factors and drivers. Here, using the NetBID2 algorithm and developing human brain RNA sequencing dataset, we identify synaptotagmin-like 3 (SYTL3) as one of the top drivers of early human brain development. Interestingly, SYTL3 exhibits high activity but low expression in both early developmental human cortex and human embryonic stem cell (hESC)-derived neurons. Knockout of SYTL3 (SYTL3-KO) in human neurons or knockdown of Sytl3 in embryonic mouse cortex markedly promotes neuronal migration. SYTL3-KO causes an abnormal distribution of deep-layer neurons in brain organoids and reduces presynaptic neurotransmitter release in hESC-derived neurons. We further demonstrate that SYTL3-KO-accelerated neuronal migration is modulated by high expression of matrix metalloproteinases. Together, based on bioinformatics and biological experiments, we identify SYTL3 as a regulator of cortical neuronal migration in human and mouse developing brains.
Collapse
Affiliation(s)
- Xinran Dong
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Lin Yang
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Kaiyi Liu
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Xiaoli Ji
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China; Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Chuanqing Tang
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Wanxing Li
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Ling Ma
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Yuting Mei
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Ting Peng
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Ban Feng
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, Shanghai, China
| | - Ziyan Wu
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, Shanghai, China
| | - Qingyuan Tang
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Yanyan Gao
- Ultrasonography Department, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Kai Yan
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Wenhao Zhou
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China; Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, Shanghai 201102, China.
| | - Man Xiong
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China.
| |
Collapse
|
41
|
Calmodulin Bidirectionally Regulates Evoked and Spontaneous Neurotransmitter Release at Retinal Ribbon Synapses. eNeuro 2021; 8:ENEURO.0257-20.2020. [PMID: 33293457 PMCID: PMC7808332 DOI: 10.1523/eneuro.0257-20.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 11/17/2020] [Accepted: 11/21/2020] [Indexed: 11/21/2022] Open
Abstract
For decades, a role for the Ca2+-binding protein calmodulin (CaM) in Ca2+-dependent presynaptic modulation of synaptic transmission has been recognized. Here, we investigated the influence of CaM on evoked and spontaneous neurotransmission at rod bipolar (RB) cell→AII amacrine cell synapses in the mouse retina. Our work was motivated by the observations that expression of CaM in RB axon terminals is extremely high and that [Ca2+] in RB terminals normally rises sufficiently to saturate endogenous buffers, making tonic CaM activation likely. Taking advantage of a model in which RBs can be stimulated by expressed channelrhodopsin-2 (ChR2) to avoid dialysis of the presynaptic terminal, we found that inhibition of CaM dramatically decreased evoked release by inhibition of presynaptic Ca channels while at the same time potentiating both Ca2+-dependent and Ca2+-independent spontaneous release. Remarkably, inhibition of myosin light chain kinase (MLCK), but not other CaM-dependent targets, mimicked the effects of CaM inhibition on evoked and spontaneous release. Importantly, initial antagonism of CaM occluded the effect of subsequent inhibition of MLCK on spontaneous release. We conclude that CaM, by acting through MLCK, bidirectionally regulates evoked and spontaneous release at retinal ribbon synapses.
Collapse
|
42
|
Alten B, Zhou Q, Shin OH, Esquivies L, Lin PY, White KI, Sun R, Chung WK, Monteggia LM, Brunger AT, Kavalali ET. Role of Aberrant Spontaneous Neurotransmission in SNAP25-Associated Encephalopathies. Neuron 2020; 109:59-72.e5. [PMID: 33147442 DOI: 10.1016/j.neuron.2020.10.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/09/2020] [Accepted: 10/07/2020] [Indexed: 01/19/2023]
Abstract
SNARE (soluble N-ethylmaleimide sensitive factor attachment protein receptor) complex, composed of synaptobrevin, syntaxin, and SNAP25, forms the essential fusion machinery for neurotransmitter release. Recent studies have reported several mutations in the gene encoding SNAP25 as a causative factor for developmental and epileptic encephalopathies of infancy and childhood with diverse clinical manifestations. However, it remains unclear how SNAP25 mutations give rise to these disorders. Here, we show that although structurally clustered mutations in SNAP25 give rise to related synaptic transmission phenotypes, specific alterations in spontaneous neurotransmitter release are a key factor to account for disease heterogeneity. Importantly, we identified a single mutation that augments spontaneous release without altering evoked release, suggesting that aberrant spontaneous release is sufficient to cause disease in humans.
Collapse
Affiliation(s)
- Baris Alten
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240-7933, USA
| | - Qiangjun Zhou
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240-7933, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240-7933, USA
| | - Ok-Ho Shin
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240-7933, USA
| | - Luis Esquivies
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Pei-Yi Lin
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240-7933, USA
| | - K Ian White
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Rong Sun
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240-7933, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240-7933, USA
| | - Wendy K Chung
- Department of Pediatrics (in Medicine), Columbia University Medical Center, New York, NY 10032, USA
| | - Lisa M Monteggia
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240-7933, USA
| | - Axel T Brunger
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Ege T Kavalali
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37240-7933, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240-7933, USA.
| |
Collapse
|
43
|
Zhou M, Melin MD, Xu W, Südhof TC. Dysfunction of parvalbumin neurons in the cerebellar nuclei produces an action tremor. J Clin Invest 2020; 130:5142-5156. [PMID: 32634124 PMCID: PMC7524475 DOI: 10.1172/jci135802] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 06/24/2020] [Indexed: 12/16/2022] Open
Abstract
Essential tremor is a common brain disorder affecting millions of people, yet the neuronal mechanisms underlying this prevalent disease remain elusive. Here, we showed that conditional deletion of synaptotagmin-2, the fastest Ca2+ sensor for synaptic neurotransmitter release, from parvalbumin neurons in mice caused an action tremor syndrome resembling the core symptom of essential tremor patients. Combining brain region-specific and cell type-specific genetic manipulation methods, we found that deletion of synaptotagmin-2 from excitatory parvalbumin-positive neurons in cerebellar nuclei was sufficient to generate an action tremor. The synaptotagmin-2 deletion converted synchronous into asynchronous neurotransmitter release in projections from cerebellar nuclei neurons onto gigantocellular reticular nucleus neurons, which might produce an action tremor by causing signal oscillations during movement. The tremor was rescued by completely blocking synaptic transmission with tetanus toxin in cerebellar nuclei, which also reversed the tremor phenotype in the traditional harmaline-induced essential tremor model. Using a promising animal model for action tremor, our results thus characterized a synaptic circuit mechanism that may underlie the prevalent essential tremor disorder.
Collapse
Affiliation(s)
- Mu Zhou
- Department of Molecular and Cellular Physiology and
| | | | - Wei Xu
- Department of Molecular and Cellular Physiology and
| | - Thomas C. Südhof
- Department of Molecular and Cellular Physiology and
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
44
|
Li L, Liu H, Hall Q, Wang W, Yu Y, Kaplan JM, Hu Z. A Hyperactive Form of unc-13 Enhances Ca 2+ Sensitivity and Synaptic Vesicle Release Probability in C. elegans. Cell Rep 2020; 28:2979-2995.e4. [PMID: 31509756 PMCID: PMC6779330 DOI: 10.1016/j.celrep.2019.08.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 04/22/2019] [Accepted: 07/31/2019] [Indexed: 11/26/2022] Open
Abstract
Munc13 proteins play several roles in regulating shortterm synaptic plasticity. However, the underlying molecular mechanisms remain largely unclear. Here we report that C. elegans UNC-13L, a Munc13-1 ortholog, has three domains that inhibit synaptic vesicle (SV) exocytosis. These include the X (sequence between C2A and C1), C1, and C2B domains. Deleting all three inhibitory domains produces a hyperactive UNC-13 (sUNC-13) that exhibits dramatically increased neurotransmitter release, Ca2+ sensitivity of release, and release probability. The vesicular pool in unc-13 mutants rescued by sUNC-13 exhibits a faster synaptic recovery and replenishment rate, demonstrating an important role of sUNC-13 in regulating synaptic plasticity. Analysis of double mutants suggests that sUNC-13 enhances tonic release by increasing the open probability of UNC-64/syntaxin-1A, whereas its effects on evoked release appear to be mediated by additional functions, presumably by further regulating the activity of the assembled soluble N-ethylmaleimide-sensitive factor activating protein receptor (SNARE) complex. Li et al. identify three domains in UNC-13L that inhibit neurotransmitter release. Removal of the three inhibitory domains produces a hyperactive UNC-13 that dramatically increases Ca2+ sensitivity and release probability of vesicle exocytosis by opening syntaxin in a highly efficient manner.
Collapse
Affiliation(s)
- Lei Li
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research (CJCADR), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Haowen Liu
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research (CJCADR), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Qi Hall
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Wei Wang
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research (CJCADR), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Yi Yu
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research (CJCADR), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Joshua M Kaplan
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA.
| | - Zhitao Hu
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research (CJCADR), The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
45
|
Huson V, Meijer M, Dekker R, Ter Veer M, Ruiter M, van Weering JR, Verhage M, Cornelisse LN. Post-tetanic potentiation lowers the energy barrier for synaptic vesicle fusion independently of Synaptotagmin-1. eLife 2020; 9:55713. [PMID: 32831174 PMCID: PMC7500951 DOI: 10.7554/elife.55713] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 08/23/2020] [Indexed: 11/13/2022] Open
Abstract
Previously, we showed that modulation of the energy barrier for synaptic vesicle fusion boosts release rates supralinearly (Schotten, 2015). Here we show that mouse hippocampal synapses employ this principle to trigger Ca2+-dependent vesicle release and post-tetanic potentiation (PTP). We assess energy barrier changes by fitting release kinetics in response to hypertonic sucrose. Mimicking activation of the C2A domain of the Ca2+-sensor Synaptotagmin-1 (Syt1), by adding a positive charge (Syt1D232N) or increasing its hydrophobicity (Syt14W), lowers the energy barrier. Removing Syt1 or impairing its release inhibitory function (Syt19Pro) increases spontaneous release without affecting the fusion barrier. Both phorbol esters and tetanic stimulation potentiate synaptic strength, and lower the energy barrier equally well in the presence and absence of Syt1. We propose a model where tetanic stimulation activates Syt1-independent mechanisms that lower the energy barrier and act additively with Syt1-dependent mechanisms to produce PTP by exerting multiplicative effects on release rates.
Collapse
Affiliation(s)
- Vincent Huson
- Department of Functional Genomics, Clinical Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center- Location VUmc, Amsterdam, Netherlands
| | - Marieke Meijer
- Department of Functional Genomics, Clinical Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center- Location VUmc, Amsterdam, Netherlands
| | - Rien Dekker
- Department of Functional Genomics, Clinical Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center- Location VUmc, Amsterdam, Netherlands
| | - Mirelle Ter Veer
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, Netherlands
| | - Marvin Ruiter
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, Netherlands
| | - Jan Rt van Weering
- Department of Functional Genomics, Clinical Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center- Location VUmc, Amsterdam, Netherlands
| | - Matthijs Verhage
- Department of Functional Genomics, Clinical Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center- Location VUmc, Amsterdam, Netherlands.,Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, Netherlands
| | - Lennart Niels Cornelisse
- Department of Functional Genomics, Clinical Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center- Location VUmc, Amsterdam, Netherlands
| |
Collapse
|
46
|
Voleti R, Jaczynska K, Rizo J. Ca 2+-dependent release of synaptotagmin-1 from the SNARE complex on phosphatidylinositol 4,5-bisphosphate-containing membranes. eLife 2020; 9:57154. [PMID: 32808925 PMCID: PMC7498268 DOI: 10.7554/elife.57154] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 08/12/2020] [Indexed: 11/13/2022] Open
Abstract
The Ca2+ sensor synaptotagmin-1 and the SNARE complex cooperate to trigger neurotransmitter release. Structural studies elucidated three distinct synaptotagmin-1-SNARE complex binding modes involving 'polybasic', 'primary' and 'tripartite' interfaces of synaptotagmin-1. We investigated these interactions using NMR and fluorescence spectroscopy. Synaptotagmin-1 binds to the SNARE complex through the polybasic and primary interfaces in solution. Ca2+-free synaptotagmin-1 binds to SNARE complexes anchored on PIP2-containing nanodiscs. R398Q/R399Q and E295A/Y338W mutations at the primary interface, which strongly impair neurotransmitter release, disrupt and enhance synaptotagmin-1-SNARE complex binding, respectively. Ca2+ induces tight binding of synaptotagmin-1 to PIP2-containing nanodiscs, disrupting synaptotagmin-1-SNARE interactions. Specific effects of mutations in the polybasic region on Ca2+-dependent synaptotagmin-1-PIP2-membrane interactions correlate with their effects on release. Our data suggest that synaptotagmin-1 binds to the SNARE complex through the primary interface and that Ca2+ releases this interaction, inducing PIP2/membrane binding and allowing cooperation between synaptotagmin-1 and the SNAREs in membrane fusion to trigger release.
Collapse
Affiliation(s)
- Rashmi Voleti
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Klaudia Jaczynska
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Josep Rizo
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
47
|
Wolfes AC, Dean C. The diversity of synaptotagmin isoforms. Curr Opin Neurobiol 2020; 63:198-209. [PMID: 32663762 DOI: 10.1016/j.conb.2020.04.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 04/07/2020] [Accepted: 04/10/2020] [Indexed: 12/20/2022]
Abstract
The synaptotagmin family of molecules is known for regulating calcium-dependent membrane fusion events. Mice and humans express 17 synaptotagmin isoforms, where most studies have focused on isoforms 1, 2, and 7, which are involved in synaptic vesicle exocytosis. Recent work has highlighted how brain function relies on additional isoforms, with roles in postsynaptic receptor endocytosis, vesicle trafficking, membrane repair, synaptic plasticity, and protection against neurodegeneration, for example, in addition to the traditional concept of synaptotagmin-mediated neurotransmitter release - in neurons as well as glia, and at different timepoints. In fact, it is not uncommon for the same isoform to feature several splice isoforms, form homo- and heterodimers, and function in different subcellular locations and cell types. This review aims to highlight the diversity of synaptotagmins, offers a concise summary of key findings on all isoforms, and discusses different ways of grouping these.
Collapse
Affiliation(s)
- Anne C Wolfes
- Department of Brain Sciences, Division of Neuroscience, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK; UK Dementia Research Institute at Imperial College, London, UK
| | - Camin Dean
- German Center for Neurodegenerative Diseases, Charité University of Medicine - Berlin, 10117 Berlin, Germany.
| |
Collapse
|
48
|
Sleiman M, Stevens DR, Chitirala P, Rettig J. Cytotoxic Granule Trafficking and Fusion in Synaptotagmin7-Deficient Cytotoxic T Lymphocytes. Front Immunol 2020; 11:1080. [PMID: 32547563 PMCID: PMC7273742 DOI: 10.3389/fimmu.2020.01080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/05/2020] [Indexed: 11/13/2022] Open
Abstract
Granules of cytotoxic T lymphocytes (CTL) are derived from the lysosomal compartment. Synaptotagmin7 (Syt7) appears to be the calcium sensor triggering fusion of lysosomes in fibroblasts. Syt7 has been proposed to control cytotoxic granule (CG) fusion in lymphocytes and mice lacking Syt7 have reduced ability to clear infections. However, fusion of CG persists in the absence of Syt7. To clarify the role of Syt7 in CTL function, we have examined the fusion of cytotoxic granules of CD8+ T-lymphocytes from Syt7 knock-out mice. We have recorded granule fusion in living CTL, using total internal reflection microscopy. Since Syt7 is considered a high affinity calcium-sensor specialized for fusion under low calcium conditions, we have compared cytotoxic granule fusion under low and high calcium conditions in the same CTL. There was no difference in latencies or numbers of fusion events per CTL under low-calcium conditions, indicating that Syt7 is not required for cytotoxic granule fusion. A deficit of fusion in Syt7 KO CTL was seen when a high-calcium solution was introduced. Expressing wild type Syt7 in Syt7 KO lymphocytes reversed this deficit, confirming its Syt7-dependence. Mutations of Syt7 which disrupt calcium binding to its C2A domain reduced the efficacy of this rescue. We counted the cytotoxic granules present at the plasma membrane to determine if the lack of fusion events in the Syt7 KO CTL was due to a lack of granules. In low calcium there were no differences in fusion events per CTL, and granule numbers were similar. In high calcium, granule number was similar though wild type CTL exhibited significantly more fusion than Syt7 KO CTL. The modest differences in granule counts do not account for the lack of fusion in high calcium in Syt7 KO CTL. In Syt7 KO CTL expressing wild type Syt7, delivery of cytotoxic granules to the plasma membrane was comparable to that of wild type CTL. Syt7 KO CTL expressing Syt7 with deficient calcium binding in the C2A domain had significantly less fusion and fewer CG at the plasma membrane. These results indicate that Syt7 is involved in trafficking of CG to the plasma membrane.
Collapse
Affiliation(s)
- Marwa Sleiman
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - David R Stevens
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Praneeth Chitirala
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Jens Rettig
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| |
Collapse
|
49
|
López-Murcia FJ, Reim K, Jahn O, Taschenberger H, Brose N. Acute Complexin Knockout Abates Spontaneous and Evoked Transmitter Release. Cell Rep 2020; 26:2521-2530.e5. [PMID: 30840877 DOI: 10.1016/j.celrep.2019.02.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/05/2018] [Accepted: 02/07/2019] [Indexed: 12/21/2022] Open
Abstract
SNARE-mediated synaptic vesicle (SV) fusion is controlled by multiple regulatory proteins that determine neurotransmitter release efficiency. Complexins are essential SNARE regulators whose mode of action is unclear, as available evidence indicates positive SV fusion facilitation and negative "fusion clamp"-like activities, with the latter occurring only in certain contexts. Because these contradictory findings likely originate in part from different experimental perturbation strategies, we attempted to resolve them by examining a conditional complexin-knockout mouse line as the most stringent genetic perturbation model available. We found that acute complexin loss after synaptogenesis in autaptic and mass-cultured hippocampal neurons reduces SV fusion probability and thus abates the rates of spontaneous, synchronous, asynchronous, and delayed transmitter release but does not affect SV priming or cause "unclamping" of spontaneous SV fusion. Thus, complexins act as facilitators of SV fusion but are dispensable for "fusion clamping" in mammalian forebrain neurons.
Collapse
Affiliation(s)
- Francisco José López-Murcia
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Kerstin Reim
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Olaf Jahn
- Proteomics Group, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; DFG-Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain, 37073 Göttingen, Germany
| | - Holger Taschenberger
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; DFG-Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain, 37073 Göttingen, Germany.
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; DFG-Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain, 37073 Göttingen, Germany.
| |
Collapse
|
50
|
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: 31] [Impact Index Per Article: 7.8] [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
|