1
|
Zhang Y, Xing Z, Dong H, Lu T, Deng Y, Li Z, Hu B, Tan A. SV2B is a crucial factor for early larval development in the silkworm, Bombyx mori. INSECT SCIENCE 2025. [PMID: 40369800 DOI: 10.1111/1744-7917.70070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/21/2025] [Accepted: 04/10/2025] [Indexed: 05/16/2025]
Abstract
Synaptic vesicle glycoprotein 2B (SV2B) gene plays a crucial role in neuromodulation and neurotransmission and is a key regulator of synaptotagmin trafficking. However, physiological functions of this gene in insects remain poorly understood. In this study, we investigated the function of the BmSV2B gene in growth and development of silkworms. Tissue expression profiling revealed that BmSV2B is highly expressed in head and midgut. A phylogenetic tree and sequence alignment demonstrated that this gene is highly conserved among lepidopteran insects. Knockout of BmSV2B using the clustered regularly interspaced small palindromic repeats (CRISPR) / CRISPR-associated nuclease 9 (Cas9) system resulted in smaller body size compared to the wild type (WT) strain. In the BmSV2B mutants, the levels of triacylglycerol were dramatically lower than that in WT. Furthermore, we found that deletion of BmSV2B extended the developmental time of larvae and led to early larval death. High-throughput RNA sequencing and quantitative real-time polymerase chain reaction analysis showed that the expression levels of juvenile hormone-degrading genes, digestive genes, 20-hydroxyecdysone -response genes and forkhead box O (FOXO) were significantly affected by the absence of BmSV2B. Taken together, BmSV2B is essential for early larval development in silkworms and could serve as a potential target for insecticides, offering a more effective approach to pest control management.
Collapse
Affiliation(s)
- Yuting Zhang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu Province, China
| | - Zhiping Xing
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu Province, China
| | - Hui Dong
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu Province, China
| | - Tao Lu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu Province, China
| | - Yuping Deng
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu Province, China
| | - Zhipeng Li
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu Province, China
| | - Bo Hu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu Province, China
| | - Anjiang Tan
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu Province, China
| |
Collapse
|
2
|
Incontro S, Musella ML, Sammari M, Di Scala C, Fantini J, Debanne D. Lipids shape brain function through ion channel and receptor modulations: physiological mechanisms and clinical perspectives. Physiol Rev 2025; 105:137-207. [PMID: 38990068 DOI: 10.1152/physrev.00004.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Lipids represent the most abundant molecular type in the brain, with a fat content of ∼60% of the dry brain weight in humans. Despite this fact, little attention has been paid to circumscribe the dynamic role of lipids in brain function and disease. Membrane lipids such as cholesterol, phosphoinositide, sphingolipids, arachidonic acid, and endocannabinoids finely regulate both synaptic receptors and ion channels that ensure critical neural functions. After a brief introduction on brain lipids and their respective properties, we review here their role in regulating synaptic function and ion channel activity, action potential propagation, neuronal development, and functional plasticity and their contribution in the development of neurological and neuropsychiatric diseases. We also provide possible directions for future research on lipid function in brain plasticity and diseases.
Collapse
Affiliation(s)
| | | | - Malika Sammari
- UNIS, INSERM, Aix-Marseille Université, Marseille, France
| | | | | | | |
Collapse
|
3
|
Hogg JA, Cousin MA. Control of Synaptotagmin-1 Trafficking by SV2A-Mechanism and Consequences for Presynaptic Function and Dysfunction. J Neurochem 2025; 169:e16308. [PMID: 39853744 PMCID: PMC11758464 DOI: 10.1111/jnc.16308] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/17/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025]
Abstract
Synaptic vesicle protein 2A (SV2A) is an abundant synaptic vesicle cargo with an as yet unconfirmed role in presynaptic function. It is also heavily implicated in epilepsy, firstly being the target of the leading anti-seizure medication levetiracetam and secondly with loss of function mutations culminating in human disease. A range of potential presynaptic functions have been proposed for SV2A; however its interaction with the calcium sensor for synchronous neurotransmitter release, synaptotagmin-1 (Syt1), has received particular attention over the past decade. In this review we will assess the evidence that the primary role of SV2A is to control the expression and localisation of Syt1 at the presynapse. This will integrate biochemical, cell biological and physiological studies where the interaction, trafficking and functional output of Syt1 is altered by SV2A. The potential for SV2A-dependent epilepsy to be a result of dysfunctional Syt1 expression and localisation is also discussed. Finally, a series of key open questions will be posed that require resolution before a definitive role for SV2A in Syt1 function in health and disease can be confirmed.
Collapse
Affiliation(s)
- James A. Hogg
- Centre for Discovery Brain Sciences, Hugh Robson Building, George Square, University of EdinburghEdinburghScotlandUK
- Simons Initiative for the Developing Brain, Hugh Robson Building, George SquareUniversity of EdinburghEdinburghScotlandUK
- Muir Maxwell Epilepsy CentreHugh Robson Building, George Square, University of EdinburghEdinburghScotlandUK
| | - Michael A. Cousin
- Centre for Discovery Brain Sciences, Hugh Robson Building, George Square, University of EdinburghEdinburghScotlandUK
- Simons Initiative for the Developing Brain, Hugh Robson Building, George SquareUniversity of EdinburghEdinburghScotlandUK
- Muir Maxwell Epilepsy CentreHugh Robson Building, George Square, University of EdinburghEdinburghScotlandUK
| |
Collapse
|
4
|
Yuan X, Li W, Yuan Y, Zhu X, Meng Y, Wu Q, Yan Q, Zhang P. Characterization of neuronal differentiation in human adipose-derived stromal cells: morphological, molecular, and ultrastructural insights. J Neurosci Methods 2024; 412:110296. [PMID: 39357604 DOI: 10.1016/j.jneumeth.2024.110296] [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: 04/26/2024] [Revised: 09/19/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVE Adipose-derived stromal cells (ADSCs) have shown promise as a potential source of neural differentiation. In this study, we investigated the morphological, molecular and ultrastructural features of ADSCs during neuronal differentiation. METHODS ADSCs were induced in vitro and their differentiation was examined at different time points. Immunocytochemical staining was performed to detect the expression of neuron-specific markers NSE and MAP-2. Immunofluorescence double labeling and Western blot detected the co-expression of presynaptic markers (CaMKII, SynCAM1, SYN) and postsynaptic markers (PSD-95, Synapsin I). Scanning electron microscopy (SEM) was performed to detect the synaptic structural features of differentiated neurons. RESULTS ADSCs showed diverse morphological features during differentiation, gradually acquiring a neuron-like spindle shape and organized arrangement. The expression of neuron-specific markers and synaptic markers peaked at 5 h of induction. Scanning electron microscopy showed polygonal protrusions of ADSC-derived neurons, and transmission electron microscopy showed characteristic ultrastructures such as nidus, synaptic vesicle-like structures, and tight junctions. CONCLUSION Our findings suggest that ADSCs differentiated for 5 h have neuronal features, including morphological, molecular, and ultrastructural resemblance to neurons, as well as the formation of synaptic structures. These insights contribute to a better understanding of ADSC-based neuronal differentiation and pave the way for future applications in regenerative medicine and neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiaodong Yuan
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China.
| | - Wen Li
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Yi Yuan
- Children's hospital of Capital institute of pediatrics, department of pediatric othopedic, Beijing 100000, China
| | - Xuhong Zhu
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Yan Meng
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Qi Wu
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Qi Yan
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Pingshu Zhang
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China.
| |
Collapse
|
5
|
Hu S, Xie Z, Wang B, Chen Y, Jing Z, Hao Y, Yao J, Wu X, Huo J, Wei A, Qin Y, Dong N, Zheng C, Song Q, Long J, Kang X, Wang C, Xu H. STED Imaging of Vesicular Endocytosis in the Synapse. Neurosci Bull 2024; 40:1379-1395. [PMID: 38976218 PMCID: PMC11365914 DOI: 10.1007/s12264-024-01254-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/08/2024] [Indexed: 07/09/2024] Open
Abstract
Endocytosis is a fundamental biological process that couples exocytosis to maintain the homeostasis of the plasma membrane and sustained neurotransmission. Super-resolution microscopy enables optical imaging of exocytosis and endocytosis in live cells and makes an essential contribution to understanding molecular mechanisms of endocytosis in neuronal somata and other types of cells. However, visualization of exo-endocytic events at the single vesicular level in a synapse with optical imaging remains a great challenge to reveal mechanisms governing the synaptic exo-endocytotic coupling. In this protocol, we describe the technical details of stimulated emission depletion (STED) imaging of synaptic endocytosis at the single-vesicle level, from sample preparation and microscopy calibration to data acquisition and analysis.
Collapse
Affiliation(s)
- Shaoqin Hu
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Department of Neurology, the First Affiliated Hospital, Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhenli Xie
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Department of Neurology, the First Affiliated Hospital, Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Bianbian Wang
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Department of Neurology, the First Affiliated Hospital, Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yang Chen
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Department of Neurology, the First Affiliated Hospital, Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zexin Jing
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Department of Neurology, the First Affiliated Hospital, Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Ying Hao
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Department of Neurology, the First Affiliated Hospital, Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jingyu Yao
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Department of Neurology, the First Affiliated Hospital, Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xuanang Wu
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Department of Neurology, the First Affiliated Hospital, Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jingxiao Huo
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Department of Neurology, the First Affiliated Hospital, Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Anqi Wei
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Department of Neurology, the First Affiliated Hospital, Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yuhao Qin
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Nan Dong
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Department of Neurology, the First Affiliated Hospital, Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Chaowen Zheng
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Department of Neurology, the First Affiliated Hospital, Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Qian Song
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Department of Neurology, the First Affiliated Hospital, Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jiangang Long
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Department of Neurology, the First Affiliated Hospital, Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xinjiang Kang
- College of Life Sciences, Liaocheng University, Liaocheng, 252059, China.
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China.
| | - Changhe Wang
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Department of Neurology, the First Affiliated Hospital, Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, 710049, China.
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China.
| | - Huadong Xu
- Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Department of Neurology, the First Affiliated Hospital, Core Facilities Sharing Platform, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
6
|
Small C, Harper C, Jiang A, Kontaxi C, Pronot M, Yak N, Malapaka A, Davenport EC, Wallis TP, Gormal RS, Joensuu M, Martínez-Mármol R, Cousin MA, Meunier FA. SV2A controls the surface nanoclustering and endocytic recruitment of Syt1 during synaptic vesicle recycling. J Neurochem 2024; 168:3188-3208. [PMID: 39091022 DOI: 10.1111/jnc.16186] [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: 02/12/2024] [Revised: 07/04/2024] [Accepted: 07/06/2024] [Indexed: 08/04/2024]
Abstract
Following exocytosis, the recapture of plasma membrane-stranded vesicular proteins into recycling synaptic vesicles (SVs) is essential for sustaining neurotransmission. Surface clustering of vesicular proteins has been proposed to act as a 'pre-assembly' mechanism for endocytosis that ensures high-fidelity retrieval of SV cargo. Here, we used single-molecule imaging to examine the nanoclustering of synaptotagmin-1 (Syt1) and synaptic vesicle protein 2A (SV2A) in hippocampal neurons. Syt1 forms surface nanoclusters through the interaction of its C2B domain with SV2A, which are sensitive to mutations in this domain (Syt1K326A/K328A) and SV2A knockdown. SV2A co-clustering with Syt1 is reduced by blocking SV2A's cognate interaction with Syt1 (SV2AT84A). Surprisingly, impairing SV2A-Syt1 nanoclustering enhanced the plasma membrane recruitment of key endocytic protein dynamin-1, causing accelerated Syt1 endocytosis, altered intracellular sorting and decreased trafficking of Syt1 to Rab5-positive endocytic compartments. Therefore, SV2A and Syt1 are segregated from the endocytic machinery in surface nanoclusters, limiting dynamin recruitment and negatively regulating Syt1 entry into recycling SVs.
Collapse
Affiliation(s)
- Christopher Small
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia
| | - Callista Harper
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Anmin Jiang
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia
| | - Christiana Kontaxi
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Marie Pronot
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Nyakuoy Yak
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia
| | - Anusha Malapaka
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia
| | - Elizabeth C Davenport
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Tristan P Wallis
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia
| | - Rachel S Gormal
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia
| | - Merja Joensuu
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia
| | - Ramón Martínez-Mármol
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia
| | - Michael A Cousin
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia
- School of Biomedical Sciences, the University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
7
|
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
|
8
|
Zhang Y, Liu J, Mao X, Fan H, Li F, Wang S, Li J, Li M, Zuo X. Reconstruction of Vesicle Assemblies with DNA Nanorulers for Resolving Heterogeneity of Vesicles in Live Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308344. [PMID: 37921116 DOI: 10.1002/adma.202308344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/26/2023] [Indexed: 11/04/2023]
Abstract
Nanoscale vesicles such as synaptic vesicles play a pivotal role in efficient interneuronal communications in vivo. However, the coexistence of single vesicle and vesicle clusters in living cells increases the heterogeneity of vesicle populations, which largely complicates the quantitative analysis of the vesicles. The high spatiotemporal monitoring of vesicle assemblies is currently incompletely resolved. Here, this work uses synthetic vesicles and DNA nanorulers to reconstruct in vitro the vesicle assemblies that mimic vesicle clusters in living cells. DNA nanorulers program the lateral distance of vesicle assemblies from 3 to 10 nm. This work uses the carbon fiber nanoelectrode (CFNE) to amperometric monitor artificial vesicle assemblies with sub-10 nm interspaces, and obtain a larger proportion of complex events. This work resolves the heterogeneity of individual vesicle release kinetics in PC12 cells with the temporal resolution down to ≈0.1 ms. This work further analyzes the aggregation state of intracellular vesicles and the exocytosis of living cells with electrochemical vesicle cytometry. The results indicate that the exocytosis of vesicle clusters is critically dependent on the size of clusters. This technology has the potential as a tool to shed light on the heterogeneity analysis of vesicle populations.
Collapse
Affiliation(s)
- Yueyue Zhang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jiangbo Liu
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xiuhai Mao
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Hongxuan Fan
- Shanghai Soong Ching Ling School, Shanghai, 201700, China
| | - Fan Li
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Shaopeng Wang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jiang Li
- Institute of Materials Biology, Shanghai University, Shanghai, 200444, China
| | - Min Li
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xiaolei Zuo
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| |
Collapse
|
9
|
Mehta N, Mondal S, Watson ET, Cui Q, Chapman ER. The juxtamembrane linker of synaptotagmin 1 regulates Ca 2+ binding via liquid-liquid phase separation. Nat Commun 2024; 15:262. [PMID: 38177243 PMCID: PMC10766989 DOI: 10.1038/s41467-023-44414-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/12/2023] [Indexed: 01/06/2024] Open
Abstract
Synaptotagmin (syt) 1, a Ca2+ sensor for synaptic vesicle exocytosis, functions in vivo as a multimer. Syt1 senses Ca2+ via tandem C2-domains that are connected to a single transmembrane domain via a juxtamembrane linker. Here, we show that this linker segment harbors a lysine-rich, intrinsically disordered region that is necessary and sufficient to mediate liquid-liquid phase separation (LLPS). Interestingly, condensate formation negatively regulates the Ca2+-sensitivity of syt1. Moreover, Ca2+ and anionic phospholipids facilitate the observed phase separation, and increases in [Ca2+]i promote the fusion of syt1 droplets in living cells. Together, these observations suggest a condensate-mediated feedback loop that serves to fine-tune the ability of syt1 to trigger release, via alterations in Ca2+ binding activity and potentially through the impact of LLPS on membrane curvature during fusion reactions. In summary, the juxtamembrane linker of syt1 emerges as a regulator of syt1 function by driving self-association via LLPS.
Collapse
Affiliation(s)
- Nikunj Mehta
- Howard Hughes Medical Institute, Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Sayantan Mondal
- Department of Chemistry, Boston University, Boston, MA, 02215, USA
| | - Emma T Watson
- Howard Hughes Medical Institute, Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Qiang Cui
- Department of Chemistry, Boston University, Boston, MA, 02215, USA
| | - Edwin R Chapman
- Howard Hughes Medical Institute, Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
10
|
Abstract
Membrane fusion and budding mediate fundamental processes like intracellular trafficking, exocytosis, and endocytosis. Fusion is thought to open a nanometer-range pore that may subsequently close or dilate irreversibly, whereas budding transforms flat membranes into vesicles. Reviewing recent breakthroughs in real-time visualization of membrane transformations well exceeding this classical view, we synthesize a new model and describe its underlying mechanistic principles and functions. Fusion involves hemi-to-full fusion, pore expansion, constriction and/or closure while fusing vesicles may shrink, enlarge, or receive another vesicle fusion; endocytosis follows exocytosis primarily by closing Ω-shaped profiles pre-formed through the flat-to-Λ-to-Ω-shape transition or formed via fusion. Calcium/SNARE-dependent fusion machinery, cytoskeleton-dependent membrane tension, osmotic pressure, calcium/dynamin-dependent fission machinery, and actin/dynamin-dependent force machinery work together to generate fusion and budding modes differing in pore status, vesicle size, speed and quantity, controls release probability, synchronization and content release rates/amounts, and underlies exo-endocytosis coupling to maintain membrane homeostasis. These transformations, underlying mechanisms, and functions may be conserved for fusion and budding in general.
Collapse
Affiliation(s)
- Ling-Gang Wu
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA.
| | - Chung Yu Chan
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| |
Collapse
|
11
|
Mehta N, Mondal S, Watson ET, Cui Q, Chapman ER. The juxtamembrane linker of synaptotagmin 1 regulates Ca 2+ binding via liquid-liquid phase separation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.11.551903. [PMID: 37609296 PMCID: PMC10441399 DOI: 10.1101/2023.08.11.551903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Synaptotagmin (syt) 1, a Ca2+ sensor for synaptic vesicle exocytosis, functions in vivo as a multimer. Syt1 senses Ca2+ via tandem C2-domains that are connected to a single transmembrane domain via a juxtamembrane linker. Here, we show that this linker segment harbors a lysine-rich, intrinsically disordered region that is necessary and sufficient to mediate liquid-liquid phase separation (LLPS). Interestingly, condensate formation negatively regulates the Ca2+-sensitivity of syt1. Moreover, Ca2+ and anionic phospholipids facilitate the observed phase separation, and increases in [Ca2+]i promote the fusion of syt1 droplets in living cells. Together, these observations suggest a condensate-mediated feedback loop that serves to fine-tune the ability of syt1 to trigger release, via alterations in Ca2+ binding activity and potentially through the impact of LLPS on membrane curvature during fusion reactions. In summary, the juxtamembrane linker of syt1 emerges as a regulator of syt1 function by driving self-association via LLPS.
Collapse
Affiliation(s)
- Nikunj Mehta
- Howard Hughes Medical Institute, Department of Neuroscience, University of Wisconsin–Madison, Madison, WI 53705, United States
| | - Sayantan Mondal
- Department of Chemistry, Boston University, Boston, MA 02215, United States
| | - Emma T. Watson
- Howard Hughes Medical Institute, Department of Neuroscience, University of Wisconsin–Madison, Madison, WI 53705, United States
| | - Qiang Cui
- Department of Chemistry, Boston University, Boston, MA 02215, United States
| | - Edwin R. Chapman
- Howard Hughes Medical Institute, Department of Neuroscience, University of Wisconsin–Madison, Madison, WI 53705, United States
| |
Collapse
|
12
|
Bolz S, Kaempf N, Puchkov D, Krauss M, Russo G, Soykan T, Schmied C, Lehmann M, Müller R, Schultz C, Perrais D, Maritzen T, Haucke V. Synaptotagmin 1-triggered lipid signaling facilitates coupling of exo- and endocytosis. Neuron 2023; 111:3765-3774.e7. [PMID: 37738980 DOI: 10.1016/j.neuron.2023.08.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/16/2023] [Accepted: 08/16/2023] [Indexed: 09/24/2023]
Abstract
Exocytosis and endocytosis are essential physiological processes and are of prime importance for brain function. Neurotransmission depends on the Ca2+-triggered exocytosis of synaptic vesicles (SVs). In neurons, exocytosis is spatiotemporally coupled to the retrieval of an equal amount of membrane and SV proteins by compensatory endocytosis. How exocytosis and endocytosis are balanced to maintain presynaptic membrane homeostasis and, thereby, sustain brain function is essentially unknown. We combine mouse genetics with optical imaging to show that the SV calcium sensor Synaptotagmin 1 couples exocytic SV fusion to the endocytic retrieval of SV membranes by promoting the local activity-dependent formation of the signaling lipid phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) at presynaptic sites. Interference with these mechanisms impairs PI(4,5)P2-triggered SV membrane retrieval but not exocytic SV fusion. Our findings demonstrate that the coupling of SV exocytosis and endocytosis involves local Synaptotagmin 1-induced lipid signaling to maintain presynaptic membrane homeostasis in central nervous system neurons.
Collapse
Affiliation(s)
- Svenja Bolz
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Natalie Kaempf
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Dmytro Puchkov
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Michael Krauss
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Giulia Russo
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Tolga Soykan
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Christopher Schmied
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Martin Lehmann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Rainer Müller
- European Molecular Biology Laboratory (EMBL), Cell Biology and Biophysics Unit, 69117 Heidelberg, Germany
| | - Carsten Schultz
- European Molecular Biology Laboratory (EMBL), Cell Biology and Biophysics Unit, 69117 Heidelberg, Germany; Department of Chemical Physiology & Biochemistry, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| | - David Perrais
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, 33000 Bordeaux, France
| | - Tanja Maritzen
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Str. 10, 13125 Berlin, Germany; Department of Nanophysiology, University of Kaiserslautern-Landau, 67663 Kaiserslautern, Germany
| | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Str. 10, 13125 Berlin, Germany; Faculty of Biology, Chemistry, Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany; NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany.
| |
Collapse
|
13
|
Courtney KC, Mandal T, Mehta N, Wu L, Li Y, Das D, Cui Q, Chapman ER. Synaptotagmin-7 outperforms synaptotagmin-1 to promote the formation of large, stable fusion pores via robust membrane penetration. Nat Commun 2023; 14:7761. [PMID: 38012142 PMCID: PMC10681989 DOI: 10.1038/s41467-023-42497-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 10/11/2023] [Indexed: 11/29/2023] Open
Abstract
Synaptotagmin-1 and synaptotagmin-7 are two prominent calcium sensors that regulate exocytosis in neuronal and neuroendocrine cells. Upon binding calcium, both proteins partially penetrate lipid bilayers that bear anionic phospholipids, but the specific underlying mechanisms that enable them to trigger exocytosis remain controversial. Here, we examine the biophysical properties of these two synaptotagmin isoforms and compare their interactions with phospholipid membranes. We discover that synaptotagmin-1-membrane interactions are greatly influenced by membrane order; tight packing of phosphatidylserine inhibits binding due to impaired membrane penetration. In contrast, synaptotagmin-7 exhibits robust membrane binding and penetration activity regardless of phospholipid acyl chain structure. Thus, synaptotagmin-7 is a super-penetrator. We exploit these observations to specifically isolate and examine the role of membrane penetration in synaptotagmin function. Using nanodisc-black lipid membrane electrophysiology, we demonstrate that membrane penetration is a critical component that underlies how synaptotagmin proteins regulate reconstituted, exocytic fusion pores in response to calcium.
Collapse
Affiliation(s)
- Kevin C Courtney
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Taraknath Mandal
- Department of Chemistry, Boston University, Boston, MA, 02215, USA
- Department of Physics, Indian Institute of Technology - Kanpur, Kanpur, 208016, India
| | - Nikunj Mehta
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Lanxi Wu
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Yueqi Li
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
- Center for Bioanalytical Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Debasis Das
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Navy Nagar, Colaba, Mumbai, 400005, India
| | - Qiang Cui
- Department of Chemistry, Boston University, Boston, MA, 02215, USA
| | - Edwin R Chapman
- Howard Hughes Medical Institute and the Department of Neuroscience, University of Wisconsin, 1111 Highland Avenue, Madison, WI, 53705, USA.
| |
Collapse
|
14
|
Timechko EE, Yakimov AM, Paramonova AI, Usoltseva AA, Utyashev NP, Ivin NO, Utyasheva AA, Yakunina AV, Kalinin VA, Dmitrenko DV. Mass Spectrometry as a Quantitative Proteomic Analysis Tool for the Search for Temporal Lobe Epilepsy Biomarkers: A Systematic Review. Int J Mol Sci 2023; 24:11130. [PMID: 37446307 DOI: 10.3390/ijms241311130] [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: 05/30/2023] [Revised: 06/25/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Temporal lobe epilepsy (TLE) is the most common form of epilepsy in adults. Tissue reorganization at the site of the epileptogenic focus is accompanied by changes in the expression patterns of protein molecules. The study of mRNA and its corresponding proteins is crucial for understanding the pathogenesis of the disease. Protein expression profiles do not always directly correlate with the levels of their transcripts; therefore, it is protein profiling that is no less important for understanding the molecular mechanisms and biological processes of TLE. The study and annotation of proteins that are statistically significantly different in patients with TLE is an approach to search for biomarkers of this disease, various stages of its development, as well as a method for searching for specific targets for the development of a further therapeutic strategy. When writing a systematic review, the following aggregators of scientific journals were used: MDPI, PubMed, ScienceDirect, Springer, and Web of Science. Scientific articles were searched using the following keywords: "proteomic", "mass-spectrometry", "protein expression", "temporal lobe epilepsy", and "biomarkers". Publications from 2003 to the present have been analyzed. Studies of brain tissues, experimental models of epilepsy, as well as biological fluids, were analyzed. For each of the groups, aberrantly expressed proteins found in various studies were isolated. Most of the studies omitted important characteristics of the studied patients, such as: duration of illness, type and response to therapy, gender, etc. Proteins that overlap across different tissue types and different studies have been highlighted: DPYSL, SYT1, STMN1, APOE, NME1, and others. The most common biological processes for them were the positive regulation of neurofibrillary tangle assembly, the regulation of amyloid fibril formation, lipoprotein catabolic process, the positive regulation of vesicle fusion, the positive regulation of oxidative stress-induced intrinsic apoptotic signaling pathway, removal of superoxide radicals, axon extension, and the regulation of actin filament depolymerization. MS-based proteomic profiling for a relevant study must accept a number of limitations, the most important of which is the need to compare different types of neurological and, in particular, epileptic disorders. Such a criterion could increase the specificity of the search work and, in the future, lead to the discovery of biomarkers for a particular disease.
Collapse
Affiliation(s)
- Elena E Timechko
- Department of Medical Genetics and Clinical Neurophysiology of Postgraduate Education, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Alexey M Yakimov
- Department of Medical Genetics and Clinical Neurophysiology of Postgraduate Education, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Anastasia I Paramonova
- Department of Medical Genetics and Clinical Neurophysiology of Postgraduate Education, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Anna A Usoltseva
- Department of Medical Genetics and Clinical Neurophysiology of Postgraduate Education, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Nikita P Utyashev
- Federal State Budgetary Institution "National Medical and Surgical Center Named after N.I. Pirogov", 105203 Moscow, Russia
| | - Nikita O Ivin
- Federal State Budgetary Institution "National Medical and Surgical Center Named after N.I. Pirogov", 105203 Moscow, Russia
| | - Anna A Utyasheva
- Federal State Budgetary Institution "National Medical and Surgical Center Named after N.I. Pirogov", 105203 Moscow, Russia
| | - Albina V Yakunina
- Department of Neurology and Neurobiology of Postgraduate Education, Samara State Medical University, 443079 Samara, Russia
| | - Vladimir A Kalinin
- Department of Neurology and Neurobiology of Postgraduate Education, Samara State Medical University, 443079 Samara, Russia
| | - Diana V Dmitrenko
- Department of Medical Genetics and Clinical Neurophysiology of Postgraduate Education, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| |
Collapse
|
15
|
Joensuu M, Syed P, Saber SH, Lanoue V, Wallis TP, Rae J, Blum A, Gormal RS, Small C, Sanders S, Jiang A, Mahrhold S, Krez N, Cousin MA, Cooper‐White R, Cooper‐White JJ, Collins BM, Parton RG, Balistreri G, Rummel A, Meunier FA. Presynaptic targeting of botulinum neurotoxin type A requires a tripartite PSG-Syt1-SV2 plasma membrane nanocluster for synaptic vesicle entry. EMBO J 2023; 42:e112095. [PMID: 37226896 PMCID: PMC10308369 DOI: 10.15252/embj.2022112095] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 04/18/2023] [Accepted: 04/28/2023] [Indexed: 05/26/2023] Open
Abstract
The unique nerve terminal targeting of botulinum neurotoxin type A (BoNT/A) is due to its capacity to bind two receptors on the neuronal plasma membrane: polysialoganglioside (PSG) and synaptic vesicle glycoprotein 2 (SV2). Whether and how PSGs and SV2 may coordinate other proteins for BoNT/A recruitment and internalization remains unknown. Here, we demonstrate that the targeted endocytosis of BoNT/A into synaptic vesicles (SVs) requires a tripartite surface nanocluster. Live-cell super-resolution imaging and electron microscopy of catalytically inactivated BoNT/A wildtype and receptor-binding-deficient mutants in cultured hippocampal neurons demonstrated that BoNT/A must bind coincidentally to a PSG and SV2 to target synaptic vesicles. We reveal that BoNT/A simultaneously interacts with a preassembled PSG-synaptotagmin-1 (Syt1) complex and SV2 on the neuronal plasma membrane, facilitating Syt1-SV2 nanoclustering that controls endocytic sorting of the toxin into synaptic vesicles. Syt1 CRISPRi knockdown suppressed BoNT/A- and BoNT/E-induced neurointoxication as quantified by SNAP-25 cleavage, suggesting that this tripartite nanocluster may be a unifying entry point for selected botulinum neurotoxins that hijack this for synaptic vesicle targeting.
Collapse
Affiliation(s)
- Merja Joensuu
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandBrisbaneQLDAustralia
| | - Parnayan Syed
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Saber H Saber
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandBrisbaneQLDAustralia
| | - Vanessa Lanoue
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Tristan P Wallis
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - James Rae
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneQLDAustralia
| | - Ailisa Blum
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Rachel S Gormal
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Christopher Small
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Shanley Sanders
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Anmin Jiang
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
| | - Stefan Mahrhold
- Institut für ToxikologieMedizinische Hochschule HannoverHannoverGermany
| | - Nadja Krez
- Institut für ToxikologieMedizinische Hochschule HannoverHannoverGermany
| | - Michael A Cousin
- Centre for Discovery Brain Sciences, Hugh Robson BuildingUniversity of EdinburghEdinburghUK
- Muir Maxwell Epilepsy CentreUniversity of EdinburghEdinburghUK
- Simons Initiative for the Developing BrainUniversity of EdinburghEdinburghUK
| | - Ruby Cooper‐White
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandBrisbaneQLDAustralia
- School of Chemical EngineeringThe University of QueenslandBrisbaneQLDAustralia
| | - Justin J Cooper‐White
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandBrisbaneQLDAustralia
- School of Chemical EngineeringThe University of QueenslandBrisbaneQLDAustralia
- UQ Centre for Stem Cell Ageing and Regenerative EngineeringThe University of QueenslandBrisbaneQLDAustralia
| | - Brett M Collins
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneQLDAustralia
| | - Robert G Parton
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneQLDAustralia
- Centre for Microscopy and MicroanalysisThe University of QueenslandBrisbaneQLDAustralia
| | - Giuseppe Balistreri
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Department of Virology, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | - Andreas Rummel
- Institut für ToxikologieMedizinische Hochschule HannoverHannoverGermany
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQLDAustralia
- School of Biomedical SciencesThe University of QueenslandBrisbaneQLDAustralia
| |
Collapse
|
16
|
Uzay B, Kavalali ET. Genetic disorders of neurotransmitter release machinery. Front Synaptic Neurosci 2023; 15:1148957. [PMID: 37066095 PMCID: PMC10102358 DOI: 10.3389/fnsyn.2023.1148957] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/10/2023] [Indexed: 04/03/2023] Open
Abstract
Synaptic neurotransmitter release is an evolutionarily conserved process that mediates rapid information transfer between neurons as well as several peripheral tissues. Release of neurotransmitters are ensured by successive events such as synaptic vesicle docking and priming that prepare synaptic vesicles for rapid fusion. These events are orchestrated by interaction of different presynaptic proteins and are regulated by presynaptic calcium. Recent studies have identified various mutations in different components of neurotransmitter release machinery resulting in aberrant neurotransmitter release, which underlie a wide spectrum of psychiatric and neurological symptoms. Here, we review how these genetic alterations in different components of the core neurotransmitter release machinery affect the information transfer between neurons and how aberrant synaptic release affects nervous system function.
Collapse
Affiliation(s)
- Burak Uzay
- Vanderbilt Brain Institute, Nashville, TN, United States
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Ege T. Kavalali
- Vanderbilt Brain Institute, Nashville, TN, United States
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
17
|
Filipović D, Costina V, Findeisen P, Inta D. Fluoxetine Enhances Synaptic Vesicle Trafficking and Energy Metabolism in the Hippocampus of Socially Isolated Rats. Int J Mol Sci 2022; 23:ijms232315351. [PMID: 36499675 PMCID: PMC9735484 DOI: 10.3390/ijms232315351] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/27/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic social isolation (CSIS)-induced alternation in synaptic and mitochondrial function of specific brain regions is associated with major depressive disorder (MDD). Despite the wide number of available medications, treating MDD remains an important challenge. Although fluoxetine (Flx) is the most frequently prescribed antidepressant, its mode of action is still unknown. To delineate affected molecular pathways of depressive-like behavior and identify potential targets upon Flx treatment, we performed a comparative proteomic analysis of hippocampal purified synaptic terminals (synaptosomes) of rats exposed to six weeks of CSIS, an animal model of depression, and/or followed by Flx treatment (lasting three weeks of six-week CSIS) to explore synaptic protein profile changes. Results showed that Flx in controls mainly induced decreased expression of proteins involved in energy metabolism and the redox system. CSIS led to increased expression of proteins that mainly participate in Ca2+/calmodulin-dependent protein kinase II (Camk2)-related neurotransmission, vesicle transport, and ubiquitination. Flx treatment of CSIS rats predominantly increased expression of proteins involved in synaptic vesicle trafficking (exocytosis and endocytosis), and energy metabolism (glycolytic and mitochondrial respiration). Overall, these Flx-regulated changes in synaptic and mitochondrial proteins of CSIS rats might be critical targets for new therapeutic development for the treatment of MDD.
Collapse
Affiliation(s)
- Dragana Filipović
- Department of Molecular Biology and Endocrinology, “VINČA”, Institute of Nuclear Sciences—National Institute of thе Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
- Correspondence: ; Tel./Fax: +381-(11)-6455-561
| | - Victor Costina
- Institute for Clinical Chemistry, Medical Faculty Mannheim of the University of Heidelberg, University Hospital Mannheim, 68159 Mannhem, Germany
| | - Peter Findeisen
- Institute for Clinical Chemistry, Medical Faculty Mannheim of the University of Heidelberg, University Hospital Mannheim, 68159 Mannhem, Germany
| | - Dragos Inta
- Department for Community Health Faculty of Natural Sciences, Medicine University of Fribourg, 1700 Fribourg, Switzerland
- Department of Biomedicine, University of Basel, 4052 Basel, Switzerland
| |
Collapse
|
18
|
Ramirez-Franco J, Azzaz F, Sangiardi M, Ferracci G, Youssouf F, Popoff MR, Seagar M, Lévêque C, Fantini J, El Far O. Molecular landscape of BoNT/B bound to a membrane-inserted synaptotagmin/ganglioside complex. Cell Mol Life Sci 2022; 79:496. [PMID: 36006520 PMCID: PMC11073447 DOI: 10.1007/s00018-022-04527-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/03/2022]
Abstract
Botulinum neurotoxin serotype B (BoNT/B) uses two separate protein and polysialoglycolipid-binding pockets to interact with synaptotagmin 1/2 and gangliosides. However, an integrated model of BoNT/B bound to its neuronal receptors in a native membrane topology is still lacking. Using a panel of in silico and experimental approaches, we present here a new model for BoNT/B binding to neuronal membranes, in which the toxin binds to a preassembled synaptotagmin-ganglioside GT1b complex and a free ganglioside allowing a lipid-binding loop of BoNT/B to interact with the glycone part of the synaptotagmin-associated GT1b. Furthermore, our data provide molecular support for the decrease in BoNT/B sensitivity in Felidae that harbor the natural variant synaptotagmin2-N59Q. These results reveal multiple interactions of BoNT/B with gangliosides and support a novel paradigm in which a toxin recognizes a protein/ganglioside complex.
Collapse
Affiliation(s)
- Jorge Ramirez-Franco
- 1INSERM, Aix-Marseille Université (AMU), UMR 1072, Unité de Neurobiologie des canaux Ioniques et de la Synapse, 13015, Marseille, France
| | - Fodil Azzaz
- 1INSERM, Aix-Marseille Université (AMU), UMR 1072, Unité de Neurobiologie des canaux Ioniques et de la Synapse, 13015, Marseille, France
| | - Marion Sangiardi
- 1INSERM, Aix-Marseille Université (AMU), UMR 1072, Unité de Neurobiologie des canaux Ioniques et de la Synapse, 13015, Marseille, France
| | - Géraldine Ferracci
- Aix-Marseille Université (AMU), CNRS, INP, Institute of Neurophysiopathology, UMR7051, PINT, PFNT, Marseille, France
| | - Fahamoe Youssouf
- 1INSERM, Aix-Marseille Université (AMU), UMR 1072, Unité de Neurobiologie des canaux Ioniques et de la Synapse, 13015, Marseille, France
| | | | - Michael Seagar
- 1INSERM, Aix-Marseille Université (AMU), UMR 1072, Unité de Neurobiologie des canaux Ioniques et de la Synapse, 13015, Marseille, France
| | - Christian Lévêque
- 1INSERM, Aix-Marseille Université (AMU), UMR 1072, Unité de Neurobiologie des canaux Ioniques et de la Synapse, 13015, Marseille, France.
| | - Jacques Fantini
- 1INSERM, Aix-Marseille Université (AMU), UMR 1072, Unité de Neurobiologie des canaux Ioniques et de la Synapse, 13015, Marseille, France
| | - Oussama El Far
- 1INSERM, Aix-Marseille Université (AMU), UMR 1072, Unité de Neurobiologie des canaux Ioniques et de la Synapse, 13015, Marseille, France.
| |
Collapse
|
19
|
Abstract
Precise and efficient coupling of endocytosis to exocytosis is critical for neurotransmission. The activity-dependent facilitation of endocytosis has been well established for efficient membrane retrieval; however, whether neural activity clamps endocytosis to avoid excessive membrane retrieval remains debatable with the mechanisms largely unknown. The present work provides compelling evidence that synaptotagmin-1 (Syt1) functions as a primary bidirectional Ca2+ sensor to promote slow, small-sized clathrin-mediated endocytosis but inhibit the fast, large-sized bulk endocytosis during elevated neural activity, the disruption of which leads to inefficient vesicle recycling under mild stimulation but excessive membrane retrieval following sustained neurotransmission. Thus, Syt1 serves as a fine-tuning Ca2+ sensor to ensure both efficient and precise coupling of endocytosis to exocytosis in response to different neural activities. Exocytosis and endocytosis are tightly coupled. In addition to initiating exocytosis, Ca2+ plays critical roles in exocytosis–endocytosis coupling in neurons and nonneuronal cells. Both positive and negative roles of Ca2+ in endocytosis have been reported; however, Ca2+ inhibition in endocytosis remains debatable with unknown mechanisms. Here, we show that synaptotagmin-1 (Syt1), the primary Ca2+ sensor initiating exocytosis, plays bidirectional and opposite roles in exocytosis–endocytosis coupling by promoting slow, small-sized clathrin-mediated endocytosis but inhibiting fast, large-sized bulk endocytosis. Ca2+-binding ability is required for Syt1 to regulate both types of endocytic pathways, the disruption of which leads to inefficient vesicle recycling under mild stimulation and excessive membrane retrieval following intense stimulation. Ca2+-dependent membrane tubulation may explain the opposite endocytic roles of Syt1 and provides a general membrane-remodeling working model for endocytosis determination. Thus, Syt1 is a primary bidirectional Ca2+ sensor facilitating clathrin-mediated endocytosis but clamping bulk endocytosis, probably by manipulating membrane curvature to ensure both efficient and precise coupling of endocytosis to exocytosis.
Collapse
|
20
|
Barber CN, Goldschmidt HL, Ma Q, Devine LR, Cole RN, Huganir RL, Raben DM. Identification of Synaptic DGKθ Interactors That Stimulate DGKθ Activity. Front Synaptic Neurosci 2022; 14:855673. [PMID: 35573662 PMCID: PMC9095502 DOI: 10.3389/fnsyn.2022.855673] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/16/2022] [Indexed: 01/16/2023] Open
Abstract
Lipids and their metabolic enzymes are a critical point of regulation for the membrane curvature required to induce membrane fusion during synaptic vesicle recycling. One such enzyme is diacylglycerol kinase θ (DGKθ), which produces phosphatidic acid (PtdOH) that generates negative membrane curvature. Synapses lacking DGKθ have significantly slower rates of endocytosis, implicating DGKθ as an endocytic regulator. Importantly, DGKθ kinase activity is required for this function. However, protein regulators of DGKθ's kinase activity in neurons have never been identified. In this study, we employed APEX2 proximity labeling and mass spectrometry to identify endogenous interactors of DGKθ in neurons and assayed their ability to modulate its kinase activity. Seven endogenous DGKθ interactors were identified and notably, synaptotagmin-1 (Syt1) increased DGKθ kinase activity 10-fold. This study is the first to validate endogenous DGKθ interactors at the mammalian synapse and suggests a coordinated role between DGKθ-produced PtdOH and Syt1 in synaptic vesicle recycling.
Collapse
Affiliation(s)
- Casey N. Barber
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, United States,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Hana L. Goldschmidt
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Qianqian Ma
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lauren R. Devine
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Robert N. Cole
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Richard L. Huganir
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Daniel M. Raben
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, United States,*Correspondence: Daniel M. Raben,
| |
Collapse
|
21
|
Benincore-Flórez E, El-Azaz J, Solarte GA, Rodríguez A, Reyes LH, Alméciga-Díaz CJ, Cardona C. Iduronate-2-sulfatase interactome: Validation by Yeast Two-Hybrid Assay. Heliyon 2022; 8:e09031. [PMID: 35284671 PMCID: PMC8913312 DOI: 10.1016/j.heliyon.2022.e09031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/08/2021] [Accepted: 02/24/2022] [Indexed: 11/25/2022] Open
Abstract
Mucopolysaccharidosis type II (MPS II), also known as Hunter syndrome, is a rare X-linked recessive disease caused by a deficiency of the lysosomal enzyme iduronate-2-sulfatase (IDS), which activates intracellular accumulation of nonmetabolized glycosaminoglycans such as heparan sulfate and dermatan sulfate. This accumulation causes severe damage to several tissues, principally the central nervous system. Previously, we identified 187 IDS-protein interactions in the mouse brain. To validate a subset of these interactions, we selected and cloned the coding regions of 10 candidate genes to perform a targeted yeast two-hybrid assay. The results allowed the identification of the physical interaction of IDS with LSAMP and SYT1. Although the physiological relevance of these complexes is unknown, recent advances allow us to point out that these interactions could be involved in vesicular trafficking of IDS through the interaction with SYT1, as well as to the ability to form a transcytosis module between the cellular components of the blood-brain-barrier (BBB) through its interaction with LSAMP. These results may shed light on the role of IDS on cellular homeostasis and may also contribute to the understanding of MPS II physiopathology and the development of novel therapeutic strategies to transport recombinant IDS through the brain endothelial cells toward the brain parenchyma.
Collapse
|
22
|
Synaptotagmin 1 oligomerization via the juxtamembrane linker regulates spontaneous and evoked neurotransmitter release. Proc Natl Acad Sci U S A 2021; 118:2113859118. [PMID: 34810248 PMCID: PMC8694047 DOI: 10.1073/pnas.2113859118] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2021] [Indexed: 11/18/2022] Open
Abstract
Synaptotagmin 1 (syt1) is a synaptic vesicle (SV) protein that is rapidly activated by Ca2+ influx into presynaptic nerve terminals, triggering SV exocytosis. Syt1 also inhibits exocytosis, prior to Ca2+ influx, and thus helps synchronize evoked exocytosis upon Ca2+ binding. Herein, we identified a cluster of lysine residues, in the oft-ignored juxtamembrane linker region of syt1, that governs homo-multimerization in an anionic lipid-dependent manner. Neutralization of this positively charged region abolished syt1 self-association on phospholipid bilayers in vitro. Subsequently, in neurons, we found mutations that disrupted syt1 self-association were correlated with defects in clamping spontaneous SV release and in triggering and synchronizing evoked exocytosis. Thus, syt1 regulates SV exocytosis as an oligomer via charged residues in the juxtamembrane linker. Synaptotagmin 1 (syt1) is a Ca2+ sensor that regulates synaptic vesicle exocytosis. Cell-based experiments suggest that syt1 functions as a multimer; however, biochemical and electron microscopy studies have yielded contradictory findings regarding putative self-association. Here, we performed dynamic light scattering on syt1 in solution, followed by electron microscopy, and we used atomic force microscopy to study syt1 self-association on supported lipid bilayers under aqueous conditions. Ring-like multimers were clearly observed. Multimerization was enhanced by Ca2+ and required anionic phospholipids. Large ring-like structures (∼180 nm) were reduced to smaller rings (∼30 nm) upon neutralization of a cluster of juxtamembrane lysine residues; further substitution of residues in the second C2-domain completely abolished self-association. When expressed in neurons, syt1 mutants with graded reductions in self-association activity exhibited concomitant reductions in 1) clamping spontaneous release and 2) triggering and synchronizing evoked release. Thus, the juxtamembrane linker of syt1 plays a crucial role in exocytosis by mediating multimerization.
Collapse
|
23
|
Liu J, Mao R, Lao IW, Yu L, Bai Q, Zhou X, Wang J. GLI1-altered mesenchymal tumor: a clinicopathological and molecular analysis of ten additional cases of an emerging entity. Virchows Arch 2021; 480:1087-1099. [PMID: 34779913 DOI: 10.1007/s00428-021-03224-0] [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: 09/13/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 12/14/2022]
Abstract
We report 10 additional cases of GLI1-altered mesenchymal tumor to further delineate its clinicopathological and molecular spectrum. There were seven males and three females with a median age of 31 years (range 1.3 ~ 75 years). Five tumors arose in the oral cavity, one each in the stomach, uterine cervix, elbow, groin, and thigh. Histologically, all cases except one were composed of monomorphic round to epithelioid cells showing an infiltrative multinodular growth pattern. The neoplastic cells were surrounded by a rich network of capillary vessels. Vessel invasion or subendothelial protrusion into the vascular space was commonly present. One tumor developed regional lymph node metastasis. The remaining case showed a predominantly spindle cell tumor. By immunohistochemistry, most tumors showed diffuse staining of CD56 (8/8) with variable expression of S100 protein (7/8). In three tumors harboring amplified genes, strong and diffuse nuclear staining of MDM2 (2/3) and CDK4 (3/3) were noted. Next-generation sequencing (NGS) studies revealed GLI1 fusions in 7 cases and GLI1 amplification in 2 cases, which were validated by fluorescence in situ hybridization (FISH) analysis in the majority of cases. One case did not show fusion gene by RNA-seq, but FISH revealed both amplification and break-apart of GLI1 gene. Follow-up information showed local recurrences in two patients. All other patients remained disease-free at the last follow-up. Our study further demonstrates that mesenchymal tumors with GLI1 alterations represent a distinctive clinicopathological entity. Although the tumor has a propensity for the tongue, it can also arise in somatic soft tissues as well as in visceral organs. Based on the characteristic morphological features and genomic profiles, we propose the term "GLI1-altered mesenchymal tumor" to describe this emerging entity.
Collapse
Affiliation(s)
- Jiahan Liu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Rongjun Mao
- Department of Pathology, Foshan Hospital of Chinese Traditional Medicine, Guangdong Province, Foshan, 528000, China
| | - I Weng Lao
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lin Yu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qianming Bai
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiaoyan Zhou
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jian Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
24
|
Vevea JD, Kusick GF, Courtney KC, Chen E, Watanabe S, Chapman ER. Synaptotagmin 7 is targeted to the axonal plasma membrane through γ-secretase processing to promote synaptic vesicle docking in mouse hippocampal neurons. eLife 2021; 10:e67261. [PMID: 34543184 PMCID: PMC8452306 DOI: 10.7554/elife.67261] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 08/27/2021] [Indexed: 12/28/2022] Open
Abstract
Synaptotagmin 7 (SYT7) has emerged as a key regulator of presynaptic function, but its localization and precise role in the synaptic vesicle cycle remain the subject of debate. Here, we used iGluSnFR to optically interrogate glutamate release, at the single-bouton level, in SYT7KO-dissociated mouse hippocampal neurons. We analyzed asynchronous release, paired-pulse facilitation, and synaptic vesicle replenishment and found that SYT7 contributes to each of these processes to different degrees. 'Zap-and-freeze' electron microscopy revealed that a loss of SYT7 diminishes docking of synaptic vesicles after a stimulus and inhibits the recovery of depleted synaptic vesicles after a stimulus train. SYT7 supports these functions from the axonal plasma membrane, where its localization and stability require both γ-secretase-mediated cleavage and palmitoylation. In summary, SYT7 is a peripheral membrane protein that controls multiple modes of synaptic vesicle (SV) exocytosis and plasticity, in part, through enhancing activity-dependent docking of SVs.
Collapse
Affiliation(s)
- Jason D Vevea
- Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
- Howard Hughes Medical InstituteMadisonUnited States
| | - Grant F Kusick
- Department of Cell Biology, Johns Hopkins University, School of MedicineBaltimoreUnited States
- Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University, School of MedicineBaltimoreUnited States
| | - Kevin C Courtney
- Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
- Howard Hughes Medical InstituteMadisonUnited States
| | - Erin Chen
- Department of Cell Biology, Johns Hopkins University, School of MedicineBaltimoreUnited States
| | - Shigeki Watanabe
- Department of Cell Biology, Johns Hopkins University, School of MedicineBaltimoreUnited States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, School of MedicineBaltimoreUnited States
| | - Edwin R Chapman
- Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
- Howard Hughes Medical InstituteMadisonUnited States
| |
Collapse
|
25
|
van Westen R, Poppinga J, Díez Arazola R, Toonen RF, Verhage M. Neuromodulator release in neurons requires two functionally redundant calcium sensors. Proc Natl Acad Sci U S A 2021; 118:e2012137118. [PMID: 33903230 PMCID: PMC8106342 DOI: 10.1073/pnas.2012137118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neuropeptides and neurotrophic factors secreted from dense core vesicles (DCVs) control many brain functions, but the calcium sensors that trigger their secretion remain unknown. Here, we show that in mouse hippocampal neurons, DCV fusion is strongly and equally reduced in synaptotagmin-1 (Syt1)- or Syt7-deficient neurons, but combined Syt1/Syt7 deficiency did not reduce fusion further. Cross-rescue, expression of Syt1 in Syt7-deficient neurons, or vice versa, completely restored fusion. Hence, both sensors are rate limiting, operating in a single pathway. Overexpression of either sensor in wild-type neurons confirmed this and increased fusion. Syt1 traveled with DCVs and was present on fusing DCVs, but Syt7 supported fusion largely from other locations. Finally, the duration of single DCV fusion events was reduced in Syt1-deficient but not Syt7-deficient neurons. In conclusion, two functionally redundant calcium sensors drive neuromodulator secretion in an expression-dependent manner. In addition, Syt1 has a unique role in regulating fusion pore duration.
Collapse
Affiliation(s)
- Rhodé van Westen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Clinical Genetics, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
| | - Josse Poppinga
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Rocío Díez Arazola
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Ruud F Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
- Department of Clinical Genetics, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
26
|
Wang QW, Lu SY, Liu YN, Chen Y, Wei H, Shen W, Chen YF, Fu CL, Wang YH, Dai A, Huang X, Gage FH, Xu Q, Yao J. Synaptotagmin-7 deficiency induces mania-like behavioral abnormalities through attenuating GluN2B activity. Proc Natl Acad Sci U S A 2020; 117:31438-31447. [PMID: 33229564 PMCID: PMC7733786 DOI: 10.1073/pnas.2016416117] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Synaptotagmin-7 (Syt7) probably plays an important role in bipolar-like behavioral abnormalities in mice; however, the underlying mechanisms for this have remained elusive. Unlike antidepressants that cause mood overcorrection in bipolar depression, N-methyl-d-aspartate receptor (NMDAR)-targeted drugs show moderate clinical efficacy, for unexplained reasons. Here we identified Syt7 single nucleotide polymorphisms (SNPs) in patients with bipolar disorder and demonstrated that mice lacking Syt7 or expressing the SNPs showed GluN2B-NMDAR dysfunction, leading to antidepressant behavioral consequences and avoidance of overcorrection by NMDAR antagonists. In human induced pluripotent stem cell (iPSC)-derived and mouse hippocampal neurons, Syt7 and GluN2B-NMDARs were localized to the peripheral synaptic region, and Syt7 triggered multiple forms of glutamate release to efficiently activate the juxtaposed GluN2B-NMDARs. Thus, while Syt7 deficiency and SNPs induced GluN2B-NMDAR dysfunction in mice, patient iPSC-derived neurons showed Syt7 deficit-induced GluN2B-NMDAR hypoactivity that was rescued by Syt7 overexpression. Therefore, Syt7 deficits induced mania-like behaviors in mice by attenuating GluN2B activity, which enabled NMDAR antagonists to avoid mood overcorrection.
Collapse
Affiliation(s)
- Qiu-Wen Wang
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Si-Yao Lu
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Yao-Nan Liu
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Yun Chen
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Hui Wei
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences and Peking Union Medical College, Neuroscience Center, Chinese Academy of Medical Sciences, 100005 Beijing, China
| | - Wei Shen
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Yan-Fen Chen
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Chong-Lei Fu
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Ying-Han Wang
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Anbang Dai
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Xuan Huang
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, 100020 Beijing, China
| | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Qi Xu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences and Peking Union Medical College, Neuroscience Center, Chinese Academy of Medical Sciences, 100005 Beijing, China;
| | - Jun Yao
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, 100084 Beijing, China;
| |
Collapse
|
27
|
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: 73] [Impact Index Per Article: 14.6] [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
|
28
|
Tanaka H, Kanatome A, Takagi S. Involvement of the synaptotagmin/stonin2 system in vesicular transport regulated by semaphorins in Caenorhabditis elegans epidermal cells. Genes Cells 2020; 25:391-401. [PMID: 32167217 DOI: 10.1111/gtc.12765] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 03/08/2020] [Indexed: 11/30/2022]
Abstract
Vesicular transport serves as an important mechanism for cell shape regulation during development. Although the semaphorin signaling molecule, a well-known regulator of axon guidance, induces endocytosis in the growth cone and the axonal transport of vertebrate neurons, the underlying molecular mechanisms remain largely unclear. Here, we show that the Caenorhabditis elegans SNT-1/synaptotagmin-UNC-41/stonin2 system, whose role in synaptic vesicle recycling in neurons has been studied extensively, is involved in semaphorin-regulated vesicular transport in larval epidermal cells. Mutations in the snt-1/unc-41 genes strongly suppressed the cell shape defects of semaphorin mutants. The null mutation in the semaphorin receptor gene, plx-1, altered the expression and localization pattern of endocytic and exocytic markers in the epidermal cells while repressing the transport of SNT-1-containing vesicles toward late endosome/lysosome pathways. Our findings suggest that the nematode semaphorins regulate the vesicular transport in epidermal cells in a manner distinct from that of vertebrate semaphorins in neurons.
Collapse
Affiliation(s)
- Hiroki Tanaka
- Division of Biological Science, Nagoya University Graduate School of Science, Nagoya, Japan
| | - Ayana Kanatome
- Division of Biological Science, Nagoya University Graduate School of Science, Nagoya, Japan
| | - Shin Takagi
- Division of Biological Science, Nagoya University Graduate School of Science, Nagoya, Japan
| |
Collapse
|
29
|
Bowers MR, Reist NE. Synaptotagmin: Mechanisms of an electrostatic switch. Neurosci Lett 2020; 722:134834. [DOI: 10.1016/j.neulet.2020.134834] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/06/2020] [Accepted: 02/09/2020] [Indexed: 02/09/2023]
|
30
|
Doc2 Proteins Are Not Required for the Increased Spontaneous Release Rate in Synaptotagmin-1-Deficient Neurons. J Neurosci 2020; 40:2606-2617. [PMID: 32098902 DOI: 10.1523/jneurosci.0309-19.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/10/2020] [Accepted: 01/23/2020] [Indexed: 11/21/2022] Open
Abstract
Regulated secretion is controlled by Ca2+ sensors with different affinities and subcellular distributions. Inactivation of Syt1 (synaptotagmin-1), the main Ca2+ sensor for synchronous neurotransmission in many neurons, enhances asynchronous and spontaneous release rates, suggesting that Syt1 inhibits other sensors with higher Ca2+ affinities and/or lower cooperativities. Such sensors could include Doc2a and Doc2b, which have been implicated in spontaneous and asynchronous neurotransmitter release and compete with Syt1 for binding SNARE complexes. Here, we tested this hypothesis using triple-knock-out mice. Inactivation of Doc2a and Doc2b in Syt1-deficient neurons did not reduce the high spontaneous release rate. Overexpression of Doc2b variants in triple-knock-out neurons reduced spontaneous release but did not rescue synchronous release. A chimeric construct in which the C2AB domain of Syt1 was substituted by that of Doc2b did not support synchronous release either. Conversely, the soluble C2AB domain of Syt1 did not affect spontaneous release. We conclude that the high spontaneous release rate in synaptotagmin-deficient neurons does not involve the binding of Doc2 proteins to Syt1 binding sites in the SNARE complex. Instead, our results suggest that the C2AB domains of Syt1 and Doc2b specifically support synchronous and spontaneous release by separate mechanisms. (Both male and female neurons were studied without sex determination.)SIGNIFICANCE STATEMENT Neurotransmission in the brain is regulated by presynaptic Ca2+ concentrations. Multiple Ca2+ sensor proteins contribute to synchronous (Syt1, Syt2), asynchronous (Syt7), and spontaneous (Doc2a/Doc2b) phases of neurotransmitter release. Genetic ablation of synchronous release was previously shown to affect other release phases, suggesting that multiple sensors may compete for similar release sites, together encoding stimulus-secretion coupling over a large range of synaptic Ca2+ concentrations. Here, we investigated the extent of functional overlap between Syt1, Doc2a, and Doc2b by reintroducing wild-type and mutant proteins in triple-knock-out neurons, and conclude that the sensors are highly specialized for different phases of release.
Collapse
|
31
|
Presenilin 1 increases association with synaptotagmin 1 during normal aging. Neurobiol Aging 2019; 86:156-161. [PMID: 31864759 PMCID: PMC7325863 DOI: 10.1016/j.neurobiolaging.2019.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/10/2019] [Accepted: 10/10/2019] [Indexed: 12/05/2022]
Abstract
Presenilin 1 (PS1), the catalytic component of gamma secretase, associates with synaptotagmin 1 (Syt-1). This interaction is decreased in the brains of patients with sporadic Alzheimer’s disease. However, it remains unclear how this interaction changes during normal aging. Because aging is a risk factor for Alzheimer’s disease, we sought to identify changes in PS1 and Syt-1 association during aging in primary neurons in vitro and mouse brain sections ex vivo. We also tested the effect of aging on the calcium dependence of the interaction by treating neurons aged in vitro with KCl. We found that PS1 and Syt-1 increase their association with age, an effect that is more robust in neuronal processes than cell bodies. Treatment with KCl triggered the interaction in both young and old neurons. Baseline calcium levels and calcium in ux in response to KCl treatment were significantly higher in older neurons, which can partially explain the increase in PS1/Syt-1 binding with age. These results suggest a compensatory mechanism during normal aging to offset detrimental age-associated effects.
Collapse
|
32
|
Varga K, Jiang ZJ, Gong LW. Phosphatidylserine is critical for vesicle fission during clathrin-mediated endocytosis. J Neurochem 2019; 152:48-60. [PMID: 31587282 DOI: 10.1111/jnc.14886] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/25/2019] [Accepted: 10/01/2019] [Indexed: 12/11/2022]
Abstract
Phosphatidylserine (PS), a negatively charged phospholipid present predominantly at the inner leaflet of the plasma membrane, has been widely implicated in many cellular processes including membrane trafficking. Along this line, PS has been demonstrated to be important for endocytosis, however, the involved mechanisms remain uncertain. By monitoring clathrin-mediated endocytosis (CME) of single vesicles in mouse chromaffin cells using cell-attached capacitance measurements that offer millisecond time resolution, we demonstrate in the present study that the fission-pore duration is reduced by PS addition, indicating a stimulatory role of PS in regulating the dynamics of vesicle fission during CME. Furthermore, our results show that the PS-mediated effect on the fission-pore duration is Ca2+ -dependent and abolished in the absence of synaptotagmin 1 (Syt1), implying that Syt1 is necessary for the stimulatory role of PS in vesicle fission during CME. Consistently, a Syt1 mutant with a defective PS-Syt1 interaction increases the fission-pore duration. Taken together, our study suggests that PS-Syt1 interaction may be critical in regulating fission dynamics during CME.
Collapse
Affiliation(s)
- Kelly Varga
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA.,Department of Biological Sciences, University of North Texas at Dallas, Dallas, Texas, USA
| | - Zhong-Jiao Jiang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Liang-Wei Gong
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
33
|
Protein Kinase C and Calmodulin Serve As Calcium Sensors for Calcium-Stimulated Endocytosis at Synapses. J Neurosci 2019; 39:9478-9490. [PMID: 31628181 DOI: 10.1523/jneurosci.0182-19.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 08/27/2019] [Accepted: 10/07/2019] [Indexed: 02/04/2023] Open
Abstract
Calcium influx triggers and facilitates endocytosis, which recycles vesicles and thus sustains synaptic transmission. Despite decades of studies, the underlying calcium sensor remained not well understood. Here, we examined two calcium binding proteins, protein kinase C (PKC) and calmodulin. Whether PKC is involved in endocytosis was unclear; whether calmodulin acts as a calcium sensor for endocytosis was neither clear, although calmodulin involvement in endocytosis had been suggested. We generated PKC (α or β-isoform) and calmodulin (calmodulin 2 gene) knock-out mice of either sex and measured endocytosis with capacitance measurements, pHluorin imaging and electron microscopy. We found that these knock-outs inhibited slow (∼10-30 s) and rapid (<∼3 s) endocytosis at large calyx-type calyces, and inhibited slow endocytosis and bulk endocytosis (forming large endosome-like structures) at small conventional hippocampal synapses, suggesting the involvement of PKC and calmodulin in three most common forms of endocytosis-the slow, rapid and bulk endocytosis. Inhibition of slow endocytosis in PKC or calmodulin 2 knock-out hippocampal synapses was rescued by overexpressing wild-type PKC or calmodulin, but not calcium-binding-deficient PKC or calmodulin mutant, respectively, suggesting that calcium stimulates endocytosis by binding with its calcium sensor PKC and calmodulin. PKC and calmodulin 2 knock-out inhibited calcium-dependent vesicle mobilization to the readily releasable pool, suggesting that PKC and calmodulin may mediate calcium-dependent facilitation of vesicle mobilization. These findings shed light on the molecular signaling link among calcium, endocytosis and vesicle mobilization that are crucial in maintaining synaptic transmission and neuronal network activity.SIGNIFICANCE STATEMENT Vesicle fusion releases neurotransmitters to mediate synaptic transmission. To sustain synaptic transmission, fused vesicles must be retrieved via endocytosis. Accumulating evidence suggests that calcium influx triggers synaptic vesicle endocytosis. However, how calcium triggers endocytosis is not well understood. Using genetic tools together with capacitance measurements, optical imaging and electron microscopy, we identified two calcium sensors, including protein kinase C (α and β isoforms) and calmodulin, for the most commonly observed forms of endocytosis: slow, rapid, and bulk. We also found that these two proteins are involved in calcium-dependent vesicle mobilization to the readily releasable pool. These results provide the molecular signaling link among calcium, endocytosis, and vesicle mobilization that are essential in sustaining synaptic transmission and neuronal network activity.
Collapse
|
34
|
Courtney NA, Bao H, Briguglio JS, Chapman ER. Synaptotagmin 1 clamps synaptic vesicle fusion in mammalian neurons independent of complexin. Nat Commun 2019; 10:4076. [PMID: 31501440 PMCID: PMC6733930 DOI: 10.1038/s41467-019-12015-w] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 08/12/2019] [Indexed: 02/01/2023] Open
Abstract
Synaptic vesicle (SV) exocytosis is mediated by SNARE proteins. Reconstituted SNAREs are constitutively active, so a major focus has been to identify fusion clamps that regulate their activity in synapses: the primary candidates are synaptotagmin (syt) 1 and complexin I/II. Syt1 is a Ca2+ sensor for SV release that binds Ca2+ via tandem C2-domains, C2A and C2B. Here, we first determined whether these C2-domains execute distinct functions. Remarkably, the C2B domain profoundly clamped all forms of SV fusion, despite synchronizing residual evoked release and rescuing the readily-releasable pool. Release was strongly enhanced by an adjacent C2A domain, and by the concurrent binding of complexin to trans-SNARE complexes. Knockdown of complexin had no impact on C2B-mediated clamping of fusion. We postulate that the C2B domain of syt1, independent of complexin, is the molecular clamp that arrests SVs prior to Ca2+-triggered fusion.
Collapse
Affiliation(s)
- Nicholas A Courtney
- Department of Neuroscience and Howard Hughes Medical Institute, University of Wisconsin-Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Huan Bao
- Department of Neuroscience and Howard Hughes Medical Institute, University of Wisconsin-Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Joseph S Briguglio
- Department of Neuroscience and Howard Hughes Medical Institute, University of Wisconsin-Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Edwin R Chapman
- Department of Neuroscience and Howard Hughes Medical Institute, University of Wisconsin-Madison, 1111 Highland Ave., Madison, WI, 53705, USA.
| |
Collapse
|
35
|
Gangliosides interact with synaptotagmin to form the high-affinity receptor complex for botulinum neurotoxin B. Proc Natl Acad Sci U S A 2019; 116:18098-18108. [PMID: 31431523 DOI: 10.1073/pnas.1908051116] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Botulinum neurotoxin type B (BoNT/B) recognizes nerve terminals by binding to 2 receptor components: a polysialoganglioside, predominantly GT1b, and synaptotagmin 1/2. It is widely thought that BoNT/B initially binds to GT1b then diffuses in the plane of the membrane to interact with synaptotagmin. We have addressed the hypothesis that a GT1b-synaptotagmin cis complex forms the BoNT/B receptor. We identified a consensus glycosphingolipid-binding motif in the extracellular juxtamembrane domain of synaptotagmins 1/2 and confirmed by Langmuir monolayer, surface plasmon resonance, and circular dichroism that GT1b interacts with synaptotagmin peptides containing this sequence, inducing α-helical structure. Molecular modeling and tryptophan fluorescence spectroscopy were consistent with the intertwining of GT1b and synaptotagmin, involving cis interactions between the oligosaccharide and ceramide moieties of GT1b and the juxtamembrane and transmembrane domains of synaptotagmin, respectively. Furthermore, a point mutation on synaptotagmin, located outside of the BoNT/B-binding segment, inhibited GT1b binding and blocked GT1b-induced potentiation of BoNT/B binding to synaptotagmin-expressing cells. Our findings are consistent with a model in which a preassembled GT1b-synaptotagmin complex constitutes the high-affinity BoNT/B receptor.
Collapse
|
36
|
Baker K, Gordon SL, Melland H, Bumbak F, Scott DJ, Jiang TJ, Owen D, Turner BJ, Boyd SG, Rossi M, Al-Raqad M, Elpeleg O, Peck D, Mancini GMS, Wilke M, Zollino M, Marangi G, Weigand H, Borggraefe I, Haack T, Stark Z, Sadedin S, Tan TY, Jiang Y, Gibbs RA, Ellingwood S, Amaral M, Kelley W, Kurian MA, Cousin MA, Raymond FL. SYT1-associated neurodevelopmental disorder: a case series. Brain 2019; 141:2576-2591. [PMID: 30107533 PMCID: PMC6113648 DOI: 10.1093/brain/awy209] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 06/19/2018] [Indexed: 12/25/2022] Open
Abstract
Synaptotagmin 1 (SYT1) is a critical mediator of fast, synchronous, calcium-dependent neurotransmitter release and also modulates synaptic vesicle endocytosis. This paper describes 11 patients with de novo heterozygous missense mutations in SYT1. All mutations alter highly conserved residues, and cluster in two regions of the SYT1 C2B domain at positions Met303 (M303K), Asp304 (D304G), Asp366 (D366E), Ile368 (I368T) and Asn371 (N371K). Phenotypic features include infantile hypotonia, congenital ophthalmic abnormalities, childhood-onset hyperkinetic movement disorders, motor stereotypies, and developmental delay varying in severity from moderate to profound. Behavioural characteristics include sleep disturbance and episodic agitation. Absence of epileptic seizures and normal orbitofrontal head circumference are important negative features. Structural MRI is unremarkable but EEG disturbance is universal, characterized by intermittent low frequency high amplitude oscillations. The functional impact of these five de novo SYT1 mutations has been assessed by expressing rat SYT1 protein containing the equivalent human variants in wild-type mouse primary hippocampal cultures. All mutant forms of SYT1 were expressed at levels approximately equal to endogenous wild-type protein, and correctly localized to nerve terminals at rest, except for SYT1M303K, which was expressed at a lower level and failed to localize at nerve terminals. Following stimulation, SYT1I368T and SYT1N371K relocalized to nerve terminals at least as efficiently as wild-type SYT1. However, SYT1D304G and SYT1D366E failed to relocalize to nerve terminals following stimulation, indicative of impairments in endocytic retrieval and trafficking of SYT1. In addition, the presence of SYT1 variants at nerve terminals induced a slowing of exocytic rate following sustained action potential stimulation. The extent of disturbance to synaptic vesicle kinetics is mirrored by the severity of the affected individuals' phenotypes, suggesting that the efficiency of SYT1-mediated neurotransmitter release is critical to cognitive development. In summary, de novo dominant SYT1 missense mutations are associated with a recognizable neurodevelopmental syndrome, and further cases can now be diagnosed based on clinical features, electrophysiological signature and mutation characteristics. Variation in phenotype severity may reflect mutation-specific impact on the diverse physiological functions of SYT1.
Collapse
Affiliation(s)
- Kate Baker
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Wellcome Trust / MRC Building, Hills Road, Cambridge, UK.,MRC Cognition and Brain Sciences Unit, 15 Chaucer Road, Cambridge, UK
| | - Sarah L Gordon
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, 30 Royal Parade, Parkville, VIC, Australia
| | - Holly Melland
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, 30 Royal Parade, Parkville, VIC, Australia
| | - Fabian Bumbak
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, 30 Royal Parade, Parkville, VIC, Australia
| | - Daniel J Scott
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, 30 Royal Parade, Parkville, VIC, Australia.,Department of Biochemistry and Molecular Biology, University of Melbourne, 30 Royal Parade, Parkville, VIC, Australia
| | - Tess J Jiang
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, 30 Royal Parade, Parkville, VIC, Australia
| | - David Owen
- Department of Clinical Biochemistry, Cambridge Institute for Medical Research, Hills Road, Cambridge, UK
| | - Bradley J Turner
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, 30 Royal Parade, Parkville, VIC, Australia
| | - Stewart G Boyd
- Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, UK
| | - Mari Rossi
- Ambry Genetics, 15 Argonaut, Aliso Viejo, CA, USA
| | - Mohammed Al-Raqad
- Department of Clinical Genetics, Queen Rania Al-Abdullah Children Hospital, King Hussein Medical Centre, Royal Medical Services, Amman, Jordan
| | - Orly Elpeleg
- Monique and Jacques Roboh Department of Genetic Research, Hadassah, Hebrew University Medical Center, Jerusalem, Israel
| | - Dawn Peck
- University of Missouri Health Care, Columbia, MO, USA
| | - Grazia M S Mancini
- Department of Clinical Genetics, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Martina Wilke
- Department of Clinical Genetics, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Marcella Zollino
- Institute of Genomic Medicine, Catholic University, A. Gemelli Foundation, Roma, Italy
| | - Giuseppe Marangi
- Institute of Genomic Medicine, Catholic University, A. Gemelli Foundation, Roma, Italy
| | - Heike Weigand
- Department of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Dr. von Hauner's Children's Hospital, University of Munich, Munich, Germany
| | - Ingo Borggraefe
- Department of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Dr. von Hauner's Children's Hospital, University of Munich, Munich, Germany
| | - Tobias Haack
- Institute of Human Genetics, Technische Universität München, Munich, Germany.,Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Zornitza Stark
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Flemington Road, Parkville VIC, Australia
| | - Simon Sadedin
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Flemington Road, Parkville VIC, Australia.,Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Tiong Yang Tan
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Flemington Road, Parkville VIC, Australia
| | - Yunyun Jiang
- Human Genome Sequencing Center, Baylor College of Medicine, Texas, USA
| | - Richard A Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Texas, USA
| | - Sara Ellingwood
- Maine Medical Partners Pediatric Specialty Care, Congress St, Portland ME, USA
| | - Michelle Amaral
- HudsonAlpha Institute for Biotechnology, 601 Genome Way NW, Huntsville, AL, USA
| | - Whitley Kelley
- HudsonAlpha Institute for Biotechnology, 601 Genome Way NW, Huntsville, AL, USA
| | - Manju A Kurian
- Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, UK
| | - Michael A Cousin
- Centre for Discovery Brain Sciences, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh, UK
| | - F Lucy Raymond
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Wellcome Trust / MRC Building, Hills Road, Cambridge, UK
| |
Collapse
|
37
|
Abstract
Synaptotagmins (Syts) are well-established primary Ca2+ sensors to initiate presynaptic neurotransmitter release. They also play critical roles in the docking, priming, and fusion steps of exocytosis, as well as the tightly coupled exo-endocytosis, in presynapses. A recent study by Awasthi and others (2019) shows that Syt3 Ca2+-dependently modulates the postsynaptic receptor endocytosis and thereby promotes the long-term depression (LTD) and the decay of long-term potentiation (LTP). This work highlights the importance of Syt3 in modulating long-term synaptic plasticity and, importantly, extends the function of Syt proteins from presynaptic neurotransmitter release to a new promising postsynaptic receptor internalization.
Collapse
Affiliation(s)
- Xuanang Wu
- Center for Mitochondrial Biology and Medicine, the Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Shaoqin Hu
- Center for Mitochondrial Biology and Medicine, the Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xinjiang Kang
- College of Life Sciences, Liaocheng University, Liaocheng, China.,Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Changhe Wang
- Center for Mitochondrial Biology and Medicine, the Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
38
|
Viral Transfer of Mini-Otoferlins Partially Restores the Fast Component of Exocytosis and Uncovers Ultrafast Endocytosis in Auditory Hair Cells of Otoferlin Knock-Out Mice. J Neurosci 2019; 39:3394-3411. [PMID: 30833506 DOI: 10.1523/jneurosci.1550-18.2018] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 10/24/2018] [Accepted: 11/02/2018] [Indexed: 01/06/2023] Open
Abstract
Transmitter release at auditory inner hair cell (IHC) ribbon synapses involves exocytosis of glutamatergic vesicles during voltage activation of L-type Cav1.3 calcium channels. At these synapses, the fast and indefatigable release of synaptic vesicles by IHCs is controlled by otoferlin, a six-C2-domain (C2-ABCDEF) protein that functions as a high-affinity Ca2+ sensor. The molecular events by which each otoferlin C2 domain contributes to the regulation of the synaptic vesicle cycle in IHCs are still incompletely understood. Here, we investigate their role using a cochlear viral cDNA transfer approach in vivo, where IHCs of mouse lacking otoferlin (Otof -/- mice of both sexes) were virally transduced with cDNAs of various mini-otoferlins. Using patch-clamp recordings and membrane capacitance measurements, we show that the viral transfer of mini-otoferlin containing C2-ACEF, C2-EF, or C2-DEF partially restores the fast exocytotic component in Otof -/- mouse IHCs. The restoration was much less efficient with C2-ACDF, underlining the importance of the C2-EF domain. None of the mini-otoferlins tested restored the sustained component of vesicle release, explaining the absence of hearing recovery. The restoration of the fast exocytotic component in the transduced Otof -/- IHCs was also associated with a recovery of Ca2+ currents with normal amplitude and fast time inactivation, confirming that the C-terminal C2 domains of otoferlin are essential for normal gating of Cav1.3 channels. Finally, the reintroduction of the mini-otoferlins C2-EF, C2-DEF, or C2-ACEF allowed us to uncover and characterize for the first time a dynamin-dependent ultrafast endocytosis in IHCs.SIGNIFICANCE STATEMENT Otoferlin, a large six-C2-domain protein, is essential for synaptic vesicle exocytosis at auditory hair cell ribbon synapses. Here, we show that the viral expression of truncated forms of otoferlin (C2-EF, C2-DEF, and C2-ACEF) can partially rescue the fast and transient release component of exocytosis in mouse hair cells lacking otoferlin, yet cannot sustain exocytosis after long repeated stimulation. Remarkably, these hair cells also display a dynamin-dependent ultrafast endocytosis. Overall, our study uncovers the pleiotropic role of otoferlin in the hair cell synaptic vesicle cycle, notably in triggering both ultrafast exocytosis and endocytosis and recruiting synaptic vesicles to the active zone.
Collapse
|
39
|
Dason JS, Allen AM, Vasquez OE, Sokolowski MB. Distinct functions of a cGMP-dependent protein kinase in nerve terminal growth and synaptic vesicle cycling. J Cell Sci 2019; 132:jcs.227165. [DOI: 10.1242/jcs.227165] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 02/26/2019] [Indexed: 01/20/2023] Open
Abstract
Sustained neurotransmission requires the tight coupling of synaptic vesicle (SV) exocytosis and endocytosis. The mechanisms underlying this coupling are poorly understood. We tested the hypothesis that a cGMP-dependent protein kinase (PKG), encoded by the foraging (for) gene in Drosophila melanogaster, is critical for this process using a for null mutant, genomic rescues, and tissue specific rescues. We uncoupled FOR's exocytic and endocytic functions in neurotransmission using a temperature-sensitive shibire mutant in conjunction with fluorescein-assisted light inactivation of FOR. We discovered a dual role for presynaptic FOR, where FOR inhibits SV exocytosis during low frequency stimulation by negatively regulating presynaptic Ca2+ levels and maintains neurotransmission during high frequency stimulation by facilitating SV endocytosis. Additionally, glial FOR negatively regulated nerve terminal growth through TGF-β signaling and this developmental effect was independent from FOR's effects on neurotransmission. Overall, FOR plays a critical role in coupling SV exocytosis and endocytosis, thereby balancing these two components to maintain sustained neurotransmission.
Collapse
Affiliation(s)
- Jeffrey S. Dason
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, M5S 3B2, Canada
- Department of Biological Sciences, University of Windsor, Windsor, Ontario, N9B 3P4, Canada
| | - Aaron M. Allen
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, M5S 3B2, Canada
- Present Address: Centre for Neural Circuits and Behaviour, University of Oxford, OX1 3SR Oxford, UK
| | - Oscar E. Vasquez
- Department of Ecology & Evolutionary Biology, University of Toronto, Toronto, Ontario, M5S 3B2, Canada
| | - Marla B. Sokolowski
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, M5S 3B2, Canada
- Department of Ecology & Evolutionary Biology, University of Toronto, Toronto, Ontario, M5S 3B2, Canada
- Child and Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, M5G 1M1, Canada
| |
Collapse
|
40
|
Awasthi A, Ramachandran B, Ahmed S, Benito E, Shinoda Y, Nitzan N, Heukamp A, Rannio S, Martens H, Barth J, Burk K, Wang YT, Fischer A, Dean C. Synaptotagmin-3 drives AMPA receptor endocytosis, depression of synapse strength, and forgetting. Science 2018; 363:science.aav1483. [PMID: 30545844 DOI: 10.1126/science.aav1483] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 11/01/2018] [Indexed: 01/01/2023]
Abstract
Forgetting is important. Without it, the relative importance of acquired memories in a changing environment is lost. We discovered that synaptotagmin-3 (Syt3) localizes to postsynaptic endocytic zones and removes AMPA receptors from synaptic plasma membranes in response to stimulation. AMPA receptor internalization, long-term depression (LTD), and decay of long-term potentiation (LTP) of synaptic strength required calcium-sensing by Syt3 and were abolished through Syt3 knockout. In spatial memory tasks, mice in which Syt3 was knocked out learned normally but exhibited a lack of forgetting. Disrupting Syt3:GluA2 binding in a wild-type background mimicked the lack of LTP decay and lack of forgetting, and these effects were occluded in the Syt3 knockout background. Our findings provide evidence for a molecular mechanism in which Syt3 internalizes AMPA receptors to depress synaptic strength and promote forgetting.
Collapse
Affiliation(s)
- Ankit Awasthi
- Trans-synaptic Signaling Group, European Neuroscience Institute, 37077 Goettingen, Germany
| | - Binu Ramachandran
- Trans-synaptic Signaling Group, European Neuroscience Institute, 37077 Goettingen, Germany
| | - Saheeb Ahmed
- Trans-synaptic Signaling Group, European Neuroscience Institute, 37077 Goettingen, Germany
| | - Eva Benito
- German Center for Neurodegenerative Disease, 37075 Goettingen, Germany.,Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, 37075 Goettingen, Germany
| | - Yo Shinoda
- Trans-synaptic Signaling Group, European Neuroscience Institute, 37077 Goettingen, Germany
| | - Noam Nitzan
- Trans-synaptic Signaling Group, European Neuroscience Institute, 37077 Goettingen, Germany
| | - Alina Heukamp
- Trans-synaptic Signaling Group, European Neuroscience Institute, 37077 Goettingen, Germany
| | - Sabine Rannio
- Trans-synaptic Signaling Group, European Neuroscience Institute, 37077 Goettingen, Germany
| | | | - Jonas Barth
- German Center for Neurodegenerative Disease, 37075 Goettingen, Germany.,Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, 37075 Goettingen, Germany
| | - Katja Burk
- Trans-synaptic Signaling Group, European Neuroscience Institute, 37077 Goettingen, Germany
| | - Yu Tian Wang
- Brain Research Center and Department of Medicine, University of British Columbia, Vancouver, BC V6T2B5, Canada
| | - Andre Fischer
- German Center for Neurodegenerative Disease, 37075 Goettingen, Germany.,Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, 37075 Goettingen, Germany
| | - Camin Dean
- Trans-synaptic Signaling Group, European Neuroscience Institute, 37077 Goettingen, Germany.
| |
Collapse
|
41
|
Song Q, Huang M, Wang B, Kang X, Wang C. Bidirectional regulation of Ca 2+ in exo-endocytosis coupling. SCIENCE CHINA-LIFE SCIENCES 2018; 61:1583-1585. [PMID: 30484064 DOI: 10.1007/s11427-018-9429-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 11/16/2018] [Indexed: 10/27/2022]
Affiliation(s)
- Qian Song
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Mingzhu Huang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.,College of Life Sciences, Liaocheng University, Liaocheng, 252059, China
| | - Bianbian Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.,College of Life Sciences, Liaocheng University, Liaocheng, 252059, China
| | - Xinjiang Kang
- College of Life Sciences, Liaocheng University, Liaocheng, 252059, China.,DOE-Key Lab of Medical Electrophysiology, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Changhe Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
42
|
McKenzie C, Spanova M, Johnson A, Kainrath S, Zheden V, Sitte HH, Janovjak H. Isolation of synaptic vesicles from genetically engineered cultured neurons. J Neurosci Methods 2018; 312:114-121. [PMID: 30496761 DOI: 10.1016/j.jneumeth.2018.11.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 11/19/2018] [Accepted: 11/20/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Synaptic vesicles (SVs) are an integral part of the neurotransmission machinery, and isolation of SVs from their host neuron is necessary to reveal their most fundamental biochemical and functional properties in in vitro assays. Isolated SVs from neurons that have been genetically engineered, e.g. to introduce genetically encoded indicators, are not readily available but would permit new insights into SV structure and function. Furthermore, it is unclear if cultured neurons can provide sufficient starting material for SV isolation procedures. NEW METHOD Here, we demonstrate an efficient ex vivo procedure to obtain functional SVs from cultured rat cortical neurons after genetic engineering with a lentivirus. RESULTS We show that ∼108 plated cortical neurons allow isolation of suitable SV amounts for functional analysis and imaging. We found that SVs isolated from cultured neurons have neurotransmitter uptake comparable to that of SVs isolated from intact cortex. Using total internal reflection fluorescence (TIRF) microscopy, we visualized an exogenous SV-targeted marker protein and demonstrated the high efficiency of SV modification. COMPARISON WITH EXISTING METHODS Obtaining SVs from genetically engineered neurons currently generally requires the availability of transgenic animals, which is constrained by technical (e.g. cost and time) and biological (e.g. developmental defects and lethality) limitations. CONCLUSIONS These results demonstrate the modification and isolation of functional SVs using cultured neurons and viral transduction. The ability to readily obtain SVs from genetically engineered neurons will permit linking in situ studies to in vitro experiments in a variety of genetic contexts.
Collapse
Affiliation(s)
- Catherine McKenzie
- Institute of Science and Technology Austria (IST Austria), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Miroslava Spanova
- Institute of Science and Technology Austria (IST Austria), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Alexander Johnson
- Institute of Science and Technology Austria (IST Austria), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Stephanie Kainrath
- Institute of Science and Technology Austria (IST Austria), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Vanessa Zheden
- Institute of Science and Technology Austria (IST Austria), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Harald H Sitte
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Waehringerstrasse 13A, 1090, Vienna, Austria
| | - Harald Janovjak
- Institute of Science and Technology Austria (IST Austria), Am Campus 1, 3400 Klosterneuburg, Austria; Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, 15 Innovation Walk, Clayton, Melbourne, VIC 3800, Australia; European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, 15 Innovation Walk, Clayton, Melbourne, VIC 3800, Australia.
| |
Collapse
|
43
|
Pangrsic T, Vogl C. Balancing presynaptic release and endocytic membrane retrieval at hair cell ribbon synapses. FEBS Lett 2018; 592:3633-3650. [PMID: 30251250 DOI: 10.1002/1873-3468.13258] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 11/07/2022]
Abstract
The timely and reliable processing of auditory and vestibular information within the inner ear requires highly sophisticated sensory transduction pathways. On a cellular level, these demands are met by hair cells, which respond to sound waves - or alterations in body positioning - by releasing glutamate-filled synaptic vesicles (SVs) from their presynaptic active zones with unprecedented speed and exquisite temporal fidelity, thereby initiating the auditory and vestibular pathways. In order to achieve this, hair cells have developed anatomical and molecular specializations, such as the characteristic and name-giving 'synaptic ribbons' - presynaptically anchored dense bodies that tether SVs prior to release - as well as other unique or unconventional synaptic proteins. The tightly orchestrated interplay between these molecular components enables not only ultrafast exocytosis, but similarly rapid and efficient compensatory endocytosis. So far, the knowledge of how endocytosis operates at hair cell ribbon synapses is limited. In this Review, we summarize recent advances in our understanding of the SV cycle and molecular anatomy of hair cell ribbon synapses, with a focus on cochlear inner hair cells.
Collapse
Affiliation(s)
- Tina Pangrsic
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, University Medical Center Göttingen, Germany
| | - Christian Vogl
- Presynaptogenesis and Intracellular Transport in Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, University Medical Center Göttingen, Germany
| |
Collapse
|
44
|
Lou X. Sensing Exocytosis and Triggering Endocytosis at Synapses: Synaptic Vesicle Exocytosis-Endocytosis Coupling. Front Cell Neurosci 2018; 12:66. [PMID: 29593500 PMCID: PMC5861208 DOI: 10.3389/fncel.2018.00066] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/26/2018] [Indexed: 12/29/2022] Open
Abstract
The intact synaptic structure is critical for information processing in neural circuits. During synaptic transmission, rapid vesicle exocytosis increases the size of never terminals and endocytosis counteracts the increase. Accumulating evidence suggests that SV exocytosis and endocytosis are tightly connected in time and space during SV recycling, and this process is essential for synaptic function and structural stability. Research in the past has illustrated the molecular details of synaptic vesicle (SV) exocytosis and endocytosis; however, the mechanisms that timely connect these two fundamental events are poorly understood at central synapses. Here we discuss recent progress in SV recycling and summarize several emerging mechanisms by which synapses can “sense” the occurrence of exocytosis and timely initiate compensatory endocytosis. They include Ca2+ sensing, SV proteins sensing, and local membrane stress sensing. In addition, the spatial organization of endocytic zones adjacent to active zones provides a structural basis for efficient coupling between SV exocytosis and endocytosis. Through linking different endocytosis pathways with SV fusion, these mechanisms ensure necessary plasticity and robustness of nerve terminals to meet diverse physiological needs.
Collapse
Affiliation(s)
- Xuelin Lou
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
45
|
Maritzen T, Haucke V. Coupling of exocytosis and endocytosis at the presynaptic active zone. Neurosci Res 2018; 127:45-52. [DOI: 10.1016/j.neures.2017.09.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 07/13/2017] [Accepted: 08/25/2017] [Indexed: 01/08/2023]
|
46
|
Wang C, Kang X, Zhou L, Chai Z, Wu Q, Huang R, Xu H, Hu M, Sun X, Sun S, Li J, Jiao R, Zuo P, Zheng L, Yue Z, Zhou Z. Synaptotagmin-11 is a critical mediator of parkin-linked neurotoxicity and Parkinson's disease-like pathology. Nat Commun 2018; 9:81. [PMID: 29311685 PMCID: PMC5758517 DOI: 10.1038/s41467-017-02593-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 12/09/2017] [Indexed: 11/17/2022] Open
Abstract
Loss-of-function mutations in Parkin are the most common causes of autosomal recessive Parkinson’s disease (PD). Many putative substrates of parkin have been reported; their pathogenic roles, however, remain obscure due to poor characterization, particularly in vivo. Here, we show that synaptotagmin-11, encoded by a PD-risk gene SYT11, is a physiological substrate of parkin and plays critical roles in mediating parkin-linked neurotoxicity. Unilateral overexpression of full-length, but not C2B-truncated, synaptotagmin-11 in the substantia nigra pars compacta (SNpc) impairs ipsilateral striatal dopamine release, causes late-onset degeneration of dopaminergic neurons, and induces progressive contralateral motor abnormalities. Mechanistically, synaptotagmin-11 impairs vesicle pool replenishment and thus dopamine release by inhibiting endocytosis. Furthermore, parkin deficiency induces synaptotagmin-11 accumulation and PD-like neurotoxicity in mouse models, which is reversed by SYT11 knockdown in the SNpc or knockout of SYT11 restricted to dopaminergic neurons. Thus, PD-like neurotoxicity induced by parkin dysfunction requires synaptotagmin-11 accumulation in SNpc dopaminergic neurons. Mutations in the parkin, an ubiquitin ligase, are linked to Parkinson’s disease. Here the authors show that synaptotagmin-11 is a parkin substrate and that its upregulation affects dopamine release, triggers degeneration, and causes motor impairment.
Collapse
Affiliation(s)
- Changhe Wang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.,Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xinjiang Kang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.,College of Life Sciences, Liaocheng University, Liaocheng, 252059, China.,Key Lab of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Li Zhou
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Zuying Chai
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Qihui Wu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Rong Huang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Huadong Xu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Meiqin Hu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Xiaoxuan Sun
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Suhua Sun
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Jie Li
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Ruiying Jiao
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Panli Zuo
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Lianghong Zheng
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Zhenyu Yue
- Departments of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Zhuan Zhou
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
| |
Collapse
|
47
|
Mori Y, Takamori S. Molecular Signatures Underlying Synaptic Vesicle Cargo Retrieval. Front Cell Neurosci 2018; 11:422. [PMID: 29379416 PMCID: PMC5770824 DOI: 10.3389/fncel.2017.00422] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 12/15/2017] [Indexed: 12/31/2022] Open
Abstract
Efficient retrieval of the synaptic vesicle (SV) membrane from the presynaptic plasma membrane, a process called endocytosis, is crucial for the fidelity of neurotransmission, particularly during sustained neural activity. Although multiple modes of endocytosis have been identified, it is clear that the efficient retrieval of the major SV cargos into newly formed SVs during any of these modes is fundamental for synaptic transmission. It is currently believed that SVs are eventually reformed via a clathrin-dependent pathway. Various adaptor proteins recognize SV cargos and link them to clathrin, ensuring the efficient retrieval of the cargos into newly formed SVs. Here, we summarize our current knowledge of the molecular signatures within individual SV cargos that underlie efficient retrieval into SV membranes, as well as discuss possible contributions of the mechanisms under physiological conditions.
Collapse
Affiliation(s)
- Yasunori Mori
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | - Shigeo Takamori
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| |
Collapse
|
48
|
Ugur B, Bao H, Stawarski M, Duraine LR, Zuo Z, Lin YQ, Neely GG, Macleod GT, Chapman ER, Bellen HJ. The Krebs Cycle Enzyme Isocitrate Dehydrogenase 3A Couples Mitochondrial Metabolism to Synaptic Transmission. Cell Rep 2017; 21:3794-3806. [PMID: 29281828 PMCID: PMC5747319 DOI: 10.1016/j.celrep.2017.12.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Revised: 11/10/2017] [Accepted: 12/01/2017] [Indexed: 10/18/2022] Open
Abstract
Neurotransmission is a tightly regulated Ca2+-dependent process. Upon Ca2+ influx, Synaptotagmin1 (Syt1) promotes fusion of synaptic vesicles (SVs) with the plasma membrane. This requires regulation at multiple levels, but the role of metabolites in SV release is unclear. Here, we uncover a role for isocitrate dehydrogenase 3a (idh3a), a Krebs cycle enzyme, in neurotransmission. Loss of idh3a leads to a reduction of the metabolite, alpha-ketoglutarate (αKG), causing defects in synaptic transmission similar to the loss of syt1. Supplementing idh3a flies with αKG suppresses these defects through an ATP or neurotransmitter-independent mechanism. Indeed, αKG, but not glutamate, enhances Syt1-dependent fusion in a reconstitution assay. αKG promotes interaction between the C2-domains of Syt1 and phospholipids. The data reveal conserved metabolic regulation of synaptic transmission via αKG. Our studies provide a synaptic role for αKG, a metabolite that has been proposed as a treatment for aging and neurodegenerative disorders.
Collapse
Affiliation(s)
- Berrak Ugur
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Huan Bao
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA; Howard Hughes Medical Institute, University of Wisconsin, Madison, WI 53705, USA
| | - Michal Stawarski
- Department of Biological Sciences and Wilkes Honors College, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Lita R Duraine
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhongyuan Zuo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yong Qi Lin
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - G Gregory Neely
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Gregory T Macleod
- Department of Biological Sciences and Wilkes Honors College, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Edwin R Chapman
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA; Howard Hughes Medical Institute, University of Wisconsin, Madison, WI 53705, USA
| | - Hugo J Bellen
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA.
| |
Collapse
|
49
|
Kaempf N, Maritzen T. Safeguards of Neurotransmission: Endocytic Adaptors as Regulators of Synaptic Vesicle Composition and Function. Front Cell Neurosci 2017; 11:320. [PMID: 29085282 PMCID: PMC5649181 DOI: 10.3389/fncel.2017.00320] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 09/26/2017] [Indexed: 11/13/2022] Open
Abstract
Communication between neurons relies on neurotransmitters which are released from synaptic vesicles (SVs) upon Ca2+ stimuli. To efficiently load neurotransmitters, sense the rise in intracellular Ca2+ and fuse with the presynaptic membrane, SVs need to be equipped with a stringently controlled set of transmembrane proteins. In fact, changes in SV protein composition quickly compromise neurotransmission and most prominently give rise to epileptic seizures. During exocytosis SVs fully collapse into the presynaptic membrane and consequently have to be replenished to sustain neurotransmission. Therefore, surface-stranded SV proteins have to be efficiently retrieved post-fusion to be used for the generation of a new set of fully functional SVs, a process in which dedicated endocytic sorting adaptors play a crucial role. The question of how the precise reformation of SVs is achieved is intimately linked to how SV membranes are retrieved. For a long time both processes were believed to be two sides of the same coin since Clathrin-mediated endocytosis (CME), the proposed predominant SV recycling mode, will jointly retrieve SV membranes and proteins. However, with the recent proposal of Clathrin-independent SV recycling pathways SV membrane retrieval and SV reformation turn into separable events. This review highlights the progress made in unraveling the molecular mechanisms mediating the high-fidelity retrieval of SV proteins and discusses how the gathered knowledge about SV protein recycling fits in with the new notions of SV membrane endocytosis.
Collapse
Affiliation(s)
- Natalie Kaempf
- Molecular Physiology and Cell Biology Section, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Tanja Maritzen
- Molecular Physiology and Cell Biology Section, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| |
Collapse
|
50
|
Shields MC, Bowers MR, Fulcer MM, Bollig MK, Rock PJ, Sutton BR, Vrailas-Mortimer AD, Lochmüller H, Whittaker RG, Horvath R, Reist NE. Drosophila studies support a role for a presynaptic synaptotagmin mutation in a human congenital myasthenic syndrome. PLoS One 2017; 12:e0184817. [PMID: 28953919 PMCID: PMC5617158 DOI: 10.1371/journal.pone.0184817] [Citation(s) in RCA: 10] [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: 12/09/2016] [Accepted: 08/31/2017] [Indexed: 12/04/2022] Open
Abstract
During chemical transmission, the function of synaptic proteins must be coordinated to efficiently release neurotransmitter. Synaptotagmin 2, the Ca2+ sensor for fast, synchronized neurotransmitter release at the human neuromuscular junction, has recently been implicated in a dominantly inherited congenital myasthenic syndrome associated with a non-progressive motor neuropathy. In one family, a proline residue within the C2B Ca2+-binding pocket of synaptotagmin is replaced by a leucine. The functional significance of this residue has not been investigated previously. Here we show that in silico modeling predicts disruption of the C2B Ca2+-binding pocket, and we examine the in vivo effects of the homologous mutation in Drosophila. When expressed in the absence of native synaptotagmin, this mutation is lethal, demonstrating for the first time that this residue plays a critical role in synaptotagmin function. To achieve expression similar to human patients, the mutation is expressed in flies carrying one copy of the wild type synaptotagmin gene. We now show that Drosophila carrying this mutation developed neurological and behavioral manifestations similar to those of human patients and provide insight into the mechanisms underlying these deficits. Our Drosophila studies support a role for this synaptotagmin point mutation in disease etiology.
Collapse
Affiliation(s)
- Mallory C. Shields
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
- Molecular, Cellular, and Integrative Neuroscience Program, Colorado State University, Fort Collins, CO, United States of America
| | - Matthew R. Bowers
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
- Molecular, Cellular, and Integrative Neuroscience Program, Colorado State University, Fort Collins, CO, United States of America
| | - McKenzie M. Fulcer
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
| | - Madelyn K. Bollig
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
| | - Patrick J. Rock
- School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, United States of America
| | - Bryan R. Sutton
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, Texas, United States of America
| | - Alysia D. Vrailas-Mortimer
- Department of Biological Sciences, University of Denver, Denver, CO, United States of America
- School of Biological Sciences, Illinois State University, Normal, IL, United States of America
| | - Hanns Lochmüller
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, NE1 3BZ, United Kingdom
| | - Roger G. Whittaker
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE2 4HH, United Kingdom
| | - Rita Horvath
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, NE1 3BZ, United Kingdom
| | - Noreen E. Reist
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
- Molecular, Cellular, and Integrative Neuroscience Program, Colorado State University, Fort Collins, CO, United States of America
- * E-mail:
| |
Collapse
|