1
|
Yay F, Yıldırım HÇ, Kuş F, Yalçın Ş. Dynamine 3 as a diagnostic and prognostic biomarker in pancreatic cancer: Implications for early detection and targeted therapy. Biomarkers 2025; 30:147-166. [PMID: 39847517 DOI: 10.1080/1354750x.2025.2458104] [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: 09/10/2024] [Accepted: 01/19/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Dynamins are defined as a group of molecules with GTPase activity. Among them, DNM3 has gained recognition in oncology for its tumor suppressor role. Based on this, the aim of this study is to investigate the effects of the DNM3 gene in patients diagnosed with pancreatic cancer using bioinformatics databases. MATERIALS AND METHODS For differential gene expression analysis, TCGA TARGET GTEx study on the UCSC Xena and GEO datasets were utilized; for the analysis of changes in gene expression according to clinical and pathological characteristics, UALCAN was employed; for Overall Survival (OS) analysis, Kaplan-Meier Plotter was used; for gene alteration analysis, cBioPortal was utilized; for immune cell infiltration analysis, Tumor Immune Estimation Resource (TIMER) and TIMER2.0 were employed; for enrichment analyses Enrichr was used; for Gene Set Correlation Enrichment Analysis Gscore was used on GSE15471; for essentiality of DNM3 gene in pancratic cancer cell lines DepMap was used; and for the detection of miRNAs, miRDB was utilized; ENCORI was used for gene-miRNA correlation and miRNA prognosis analyses. RESULTS In the pancreatic adenocarcinoma (PAAD) cohort, DNM3 gene expression was higher in tumor samples, and there was no significant difference in expression among cancer stages. High levels of DNM3 gene expression were associated with longer OS in PAAD. A weak positive correlation was observed between DNM3 gene expression and B-Cell and CD4+ T Cell infiltrations, while a moderate positive correlation was found with CD8+ T Cell, Macrophage, Neutrophil, and Dendritic Cell infiltrations in TIMER. NK cell by QUANTISEQ, CD 4+ T Cell by TIMER, T cell regulatory (Tregs) by CIBERSORT-ABS infiltrations were positively associated with DNM3 gene expression and decreased risk in prognosis. Common lymphoid progenitor by XCELL and MDSC by TIDE infiltrations were negatively associated with DNM3 gene expression and increased risk of prognosis. Macrophage M1 by QUANTISEQ was positively associated with DNM3 gene expression and increased risk in prognosis. DNM3 gene appears to be associated with various pathways related to inflammation and the immune system. Amplification of the DNM3 gene was detected in 5 out of 175 patients. Enrichment was observed in pathways such as bacterial invasion of epithelial cells, endocytosis, endocrine and other factor-regulated calcium reabsorption, synaptic vesicle cycle, and phospholipase D signaling pathway. According to Gscore, DNM3 gene was associated with Fc epsilon RI signaling pathway, HALLMARK MTORC1 SIGNALING, HALLMARK EPITHELIAL MESENCHYMAL TRANSITION gene sets. According to ENCORI, DNM3 gene was negatively correlated with hsa-miR-203a-3p and increased expression of this miRNA was associated with adverse prognosis in PAAD. CONCLUSIONS The DNM3 gene may play a tumor suppressor role in pancreatic cancer, similar to its role in other malignancies. The contribution of immune cells may also be significant in this effect. However, in vitro studies are needed to elucidate the mechanisms triggered in pancreatic cancer.
Collapse
Affiliation(s)
- Fatih Yay
- Clinical Biochemistry Laboratory, Nigde Omer Halisdemir University Training and Research Hospital, Nigde, Turkey
| | - Hasan Çağrı Yıldırım
- Department of Medical Oncology, Nigde Omer Halisdemir University Training and Research Hospital, Nigde, Turkey
| | - Fatih Kuş
- Faculty of Medicine, Deparment of Medical Oncology, Hacettepe University, Ankara, Turkey
| | - Şuayib Yalçın
- Faculty of Medicine, Deparment of Medical Oncology, Hacettepe University, Ankara, Turkey
| |
Collapse
|
2
|
Kravčenko U, Ruwolt M, Kroll J, Yushkevich A, Zenkner M, Ruta J, Lotfy R, Wanker EE, Rosenmund C, Liu F, Kudryashev M. Molecular architecture of synaptic vesicles. Proc Natl Acad Sci U S A 2024; 121:e2407375121. [PMID: 39602275 PMCID: PMC11626200 DOI: 10.1073/pnas.2407375121] [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: 04/15/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Synaptic vesicles (SVs) store and transport neurotransmitters to the presynaptic active zone for release by exocytosis. After release, SV proteins and excess membrane are recycled via endocytosis, and new SVs can be formed in a clathrin-dependent manner. This process maintains complex molecular composition of SVs through multiple recycling rounds. Previous studies explored the molecular composition of SVs through proteomic analysis and fluorescent microscopy, proposing a model for an average SV (1). However, the structural heterogeneity and molecular architecture of individual SVs are not well described. Here, we used cryoelectron tomography to visualize molecular details of SVs isolated from mouse brains and inside cultured neurons. We describe several classes of small proteins on the SV surface and long proteinaceous densities inside SVs. We identified V-ATPases, determined a structure using subtomogram averaging, and showed them forming a complex with the membrane-embedded protein synaptophysin (Syp). Our bioluminescence assay revealed pairwise interactions between vesicle-associated membrane protein 2 and Syp and V-ATPase Voe1 domains. Interestingly, V-ATPases were randomly distributed on the surface of SVs irrespective of vesicle size. A subpopulation of isolated vesicles and vesicles inside neurons contained a partially assembled clathrin coat with an icosahedral symmetry. We observed V-ATPases under clathrin cages in several isolated clathrin-coated vesicles (CCVs). Additionally, from isolated SV preparations and within hippocampal neurons we identified clathrin baskets without vesicles. We determined their and CCVs preferential location in proximity to the cell membrane. Our analysis advances the understanding of individual SVs' diversity and their molecular architecture.
Collapse
Affiliation(s)
- Uljana Kravčenko
- In situ Structural Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin13125, Germany
- Department of Biology, Humboldt University of Berlin, Berlin, Germany
| | - Max Ruwolt
- Leibniz Research Institute for Molecular Pharmacology, Berlin, Germany
| | - Jana Kroll
- Structural Biology of Membrane-Associated Processes, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute of Chemistry and Biochemistry, Free University of Berlin, Berlin, Germany
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Artsemi Yushkevich
- In situ Structural Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin13125, Germany
- Department of Physics, Humboldt University of Berlin, Berlin, Germany
| | - Martina Zenkner
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Julia Ruta
- In situ Structural Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin13125, Germany
- Leibniz Research Institute for Molecular Pharmacology, Berlin, Germany
- Institute of Chemistry and Biochemistry, Free University of Berlin, Berlin, Germany
| | - Rowaa Lotfy
- In situ Structural Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin13125, Germany
- Institute of Pharmacy, Free University of Berlin, Berlin, Germany
| | - Erich E. Wanker
- Neuroproteomics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Christian Rosenmund
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Fan Liu
- Leibniz Research Institute for Molecular Pharmacology, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Mikhail Kudryashev
- In situ Structural Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin13125, Germany
- Institute of Medical Physics and Biophysics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
3
|
Bolz S, Haucke V. Biogenesis and reformation of synaptic vesicles. J Physiol 2024. [PMID: 39367867 DOI: 10.1113/jp286554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/12/2024] [Indexed: 10/07/2024] Open
Abstract
Communication within the nervous system relies on the calcium-triggered release of neurotransmitter molecules by exocytosis of synaptic vesicles (SVs) at defined active zone release sites. While decades of research have provided detailed insight into the molecular machinery for SV fusion, much less is known about the mechanisms that form functional SVs during the development of synapses and that control local SV reformation following exocytosis in the mature nervous system. Here we review the current state of knowledge in the field, focusing on the pathways implicated in the formation and axonal transport of SV precursor organelles and the mechanisms involved in the local reformation of SVs within nerve terminals in mature neurons. We discuss open questions and outline perspectives for future research.
Collapse
Affiliation(s)
- Svenja Bolz
- Molecular Pharmacology and Cell Biology, Leibniz Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Volker Haucke
- Molecular Pharmacology and Cell Biology, Leibniz Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
- Department of Biology, Chemistry, Pharmacy, Freie Universität Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
4
|
Alimohamadi H, Luo EWC, Gupta S, de Anda J, Yang R, Mandal T, Wong GCL. Comparing Multifunctional Viral and Eukaryotic Proteins for Generating Scission Necks in Membranes. ACS NANO 2024; 18:15545-15556. [PMID: 38838261 PMCID: PMC11846687 DOI: 10.1021/acsnano.4c00277] [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] [Indexed: 06/07/2024]
Abstract
Deterministic formation of membrane scission necks by protein machinery with multiplexed functions is critical in biology. A microbial example is M2 viroporin, a proton pump from the influenza A virus that is multiplexed with membrane remodeling activity to induce budding and scission in the host membrane during viral maturation. In comparison, the dynamin family constitutes a class of eukaryotic proteins implicated in mitochondrial fission, as well as various budding and endocytosis pathways. In the case of Dnm1, the mitochondrial fission protein in yeast, the membrane remodeling activity is multiplexed with mechanoenzyme activity to create fission necks. It is not clear why these functions are combined in these scission processes, which occur in drastically different compositions and solution conditions. In general, direct experimental access to changing neck sizes induced by individual proteins or peptide fragments is challenging due to the nanoscale dimensions and influence of thermal fluctuations. Here, we use a mechanical model to estimate the size of scission necks by leveraging small-angle X-ray scattering structural data of protein-lipid systems under different conditions. The influence of interfacial tension, lipid composition, and membrane budding morphology on the size of the induced scission necks is systematically investigated using our data and molecular dynamic simulations. We find that the M2 budding protein from the influenza A virus has robust pH-dependent membrane activity that induces nanoscopic necks within the range of spontaneous hemifission for a broad range of lipid compositions. In contrast, the sizes of scission necks generated by mitochondrial fission proteins strongly depend on lipid composition, which suggests a role for mechanical constriction.
Collapse
Affiliation(s)
- Haleh Alimohamadi
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California 90025, United States
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Elizabeth Wei-Chia Luo
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California 90025, United States
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Shivam Gupta
- Department of Physics, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Jaime de Anda
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California 90025, United States
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Rena Yang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California 90025, United States
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Taraknath Mandal
- Department of Physics, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Gerard C L Wong
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California 90025, United States
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
5
|
Wei L, Guo X, Haimov E, Obashi K, Lee SH, Shin W, Sun M, Chan CY, Sheng J, Zhang Z, Mohseni A, Ghosh Dastidar S, Wu XS, Wang X, Han S, Arpino G, Shi B, Molakarimi M, Matthias J, Wurm CA, Gan L, Taraska JW, Kozlov MM, Wu LG. Clathrin mediates membrane fission and budding by constricting membrane pores. Cell Discov 2024; 10:62. [PMID: 38862506 PMCID: PMC11166961 DOI: 10.1038/s41421-024-00677-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 04/04/2024] [Indexed: 06/13/2024] Open
Abstract
Membrane budding, which underlies fundamental processes like endocytosis, intracellular trafficking, and viral infection, is thought to involve membrane coat-forming proteins, including the most observed clathrin, to form Ω-shape profiles and helix-forming proteins like dynamin to constrict Ω-profiles' pores and thus mediate fission. Challenging this fundamental concept, we report that polymerized clathrin is required for Ω-profiles' pore closure and that clathrin around Ω-profiles' base/pore region mediates pore constriction/closure in neuroendocrine chromaffin cells. Mathematical modeling suggests that clathrin polymerization at Ω-profiles' base/pore region generates forces from its intrinsically curved shape to constrict/close the pore. This new fission function may exert broader impacts than clathrin's well-known coat-forming function during clathrin (coat)-dependent endocytosis, because it underlies not only clathrin (coat)-dependent endocytosis, but also diverse endocytic modes, including ultrafast, fast, slow, bulk, and overshoot endocytosis previously considered clathrin (coat)-independent in chromaffin cells. It mediates kiss-and-run fusion (fusion pore closure) previously considered bona fide clathrin-independent, and limits the vesicular content release rate. Furthermore, analogous to results in chromaffin cells, we found that clathrin is essential for fast and slow endocytosis at hippocampal synapses where clathrin was previously considered dispensable, suggesting clathrin in mediating synaptic vesicle endocytosis and fission. These results suggest that clathrin and likely other intrinsically curved coat proteins are a new class of fission proteins underlying vesicle budding and fusion. The half-a-century concept and studies that attribute vesicle-coat contents' function to Ω-profile formation and classify budding as coat-protein (e.g., clathrin)-dependent or -independent may need to be re-defined and re-examined by considering clathrin's pivotal role in pore constriction/closure.
Collapse
Affiliation(s)
- Lisi Wei
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Xiaoli Guo
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Ehud Haimov
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel
| | - Kazuki Obashi
- Biochemistry and Biophysics Center, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | - Sung Hoon Lee
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
- Chung-Ang University, Seoul, Republic of Korea
| | - Wonchul Shin
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Min Sun
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Chung Yu Chan
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Jiansong Sheng
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
- 900 Clopper Rd, Suite, 130, Gaithersburg, MD, USA
| | - Zhen Zhang
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
- Center of Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Ammar Mohseni
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | | | - Xin-Sheng Wu
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Xin Wang
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Sue Han
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Gianvito Arpino
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
- Emme 3 Srl - Via Luigi Meraviglia, 31 - 20020, Lainate, MI, Italy
| | - Bo Shi
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Maryam Molakarimi
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | | | | | - Lin Gan
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Justin W Taraska
- Biochemistry and Biophysics Center, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | - Michael M Kozlov
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel.
| | - Ling-Gang Wu
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA.
| |
Collapse
|
6
|
Alimohamadi H, Luo EWC, Gupta S, de Anda J, Yang R, Mandal T, Wong GCL. Comparing multifunctional viral and eukaryotic proteins for generating scission necks in membranes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.05.574447. [PMID: 38260291 PMCID: PMC10802413 DOI: 10.1101/2024.01.05.574447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Deterministic formation of membrane scission necks by protein machinery with multiplexed functions is critical in biology. A microbial example is the M2 viroporin, a proton pump from the influenza A virus which is multiplexed with membrane remodeling activity to induce budding and scission in the host membrane during viral maturation. In comparison, the dynamin family constitutes a class of eukaryotic proteins implicated in mitochondrial fission, as well as various budding and endocytosis pathways. In the case of Dnm1, the mitochondrial fission protein in yeast, the membrane remodeling activity is multiplexed with mechanoenzyme activity to create fission necks. It is not clear why these functions are combined in these scission processes, which occur in drastically different compositions and solution conditions. In general, direct experimental access to changing neck sizes induced by individual proteins or peptide fragments is challenging due to the nanoscale dimensions and influence of thermal fluctuations. Here, we use a mechanical model to estimate the size of scission necks by leveraging Small-Angle X-ray Scattering (SAXS) structural data of protein-lipid systems under different conditions. The influence of interfacial tension, lipid composition, and membrane budding morphology on the size of the induced scission necks is systematically investigated using our data and molecular dynamic simulations. We find that the M2 budding protein from the influenza A virus has robust pH-dependent membrane activity that induces nanoscopic necks within the range of spontaneous hemi-fission for a broad range of lipid compositions. In contrast, the sizes of scission necks generated by mitochondrial fission proteins strongly depend on lipid composition, which suggests a role for mechanical constriction.
Collapse
|
7
|
Xu H, Oses-Prieto JA, Khvotchev M, Jain S, Liang J, Burlingame A, Edwards RH. Adaptor protein AP-3 produces synaptic vesicles that release at high frequency by recruiting phospholipid flippase ATP8A1. Nat Neurosci 2023; 26:1685-1700. [PMID: 37723322 DOI: 10.1038/s41593-023-01434-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 08/14/2023] [Indexed: 09/20/2023]
Abstract
Neural systems encode information in the frequency of action potentials, which is then decoded by synaptic transmission. However, the rapid, synchronous release of neurotransmitters depletes synaptic vesicles (SVs), limiting release at high firing rates. How then do synapses convey information about frequency? Here, we show in mouse hippocampal neurons and slices that the adaptor protein AP-3 makes a subset of SVs that respond specifically to high-frequency stimulation. Neurotransmitter transporters slot onto these SVs in different proportions, contributing to the distinct properties of release observed at different excitatory synapses. Proteomics reveals that AP-3 targets the phospholipid flippase ATP8A1 to SVs; loss of ATP8A1 recapitulates the defect in SV mobilization at high frequency observed with loss of AP-3. The mechanism involves recruitment of synapsin by the cytoplasmically oriented phosphatidylserine translocated by ATP8A1. Thus, ATP8A1 enables the subset of SVs made by AP-3 to release at high frequency.
Collapse
Affiliation(s)
- Hongfei Xu
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Juan A Oses-Prieto
- Department of Pharmaceutical Chemistry, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Mikhail Khvotchev
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Shweta Jain
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Jocelyn Liang
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Alma Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Robert H Edwards
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA.
| |
Collapse
|
8
|
Vargas KJ, Colosi PL, Girardi E, Park JM, Harmon LE, Chandra SS. α-Synuclein colocalizes with AP180 and affects the size of clathrin lattices. J Biol Chem 2023; 299:105091. [PMID: 37516240 PMCID: PMC10470054 DOI: 10.1016/j.jbc.2023.105091] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 07/11/2023] [Accepted: 07/11/2023] [Indexed: 07/31/2023] Open
Abstract
α-Synuclein and family members β- and γ-synuclein are presynaptic proteins that sense and generate membrane curvature, properties important for synaptic vesicle (SV) cycling. αβγ-synuclein triple knockout neurons exhibit SV endocytosis deficits. Here, we investigated if α-synuclein affects clathrin assembly in vitro. Visualizing clathrin assembly on membranes using a lipid monolayer system revealed that α-synuclein increases clathrin lattices size and curvature. On cell membranes, we observe that α-synuclein is colocalized with clathrin and its adapter AP180 in a concentric ring pattern. Clathrin puncta that contain both α-synuclein and AP180 were significantly larger than clathrin puncta containing either protein alone. We determined that this effect occurs in part through colocalization of α-synuclein with the phospholipid PI(4,5)P2 in the membrane. Immuno-electron microscopy (EM) of synaptosomes uncovered that α-synuclein relocalizes from SVs to the presynaptic membrane upon stimulation, positioning α-synuclein to function on presynaptic membranes during or after stimulation. Additionally, we show that deletion of synucleins impacts brain-derived clathrin-coated vesicle size. Thus, α-synuclein affects the size and curvature of clathrin structures on membranes and functions as an endocytic accessory protein.
Collapse
Affiliation(s)
- Karina J Vargas
- Departments of Neurology and Neuroscience, Yale University, New Haven, Connecticut, USA; Marine Biological Laboratory, Woods Hole, Massachusetts, USA; Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - P L Colosi
- Departments of Neurology and Neuroscience, Yale University, New Haven, Connecticut, USA; PREP Program, Yale University, New Haven, Connecticut, USA
| | - Eric Girardi
- Departments of Neurology and Neuroscience, Yale University, New Haven, Connecticut, USA
| | - Jae-Min Park
- Departments of Neurology and Neuroscience, Yale University, New Haven, Connecticut, USA
| | - Leah E Harmon
- Departments of Neurology and Neuroscience, Yale University, New Haven, Connecticut, USA
| | - Sreeganga S Chandra
- Departments of Neurology and Neuroscience, Yale University, New Haven, Connecticut, USA.
| |
Collapse
|
9
|
Heitmann T, Barrow JC. The Role of Inositol Hexakisphosphate Kinase in the Central Nervous System. Biomolecules 2023; 13:1317. [PMID: 37759717 PMCID: PMC10526494 DOI: 10.3390/biom13091317] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/23/2023] [Accepted: 08/26/2023] [Indexed: 09/29/2023] Open
Abstract
Inositol is a unique biological small molecule that can be phosphorylated or even further pyrophosphorylated on each of its six hydroxyl groups. These numerous phosphorylation states of inositol along with the kinases and phosphatases that interconvert them comprise the inositol phosphate signaling pathway. Inositol hexakisphosphate kinases, or IP6Ks, convert the fully mono-phosphorylated inositol to the pyrophosphate 5-IP7 (also denoted IP7). There are three isoforms of IP6K: IP6K1, 2, and 3. Decades of work have established a central role for IP6Ks in cell signaling. Genetic and pharmacologic manipulation of IP6Ks in vivo and in vitro has shown their importance in metabolic disease, chronic kidney disease, insulin signaling, phosphate homeostasis, and numerous other cellular and physiologic processes. In addition to these peripheral processes, a growing body of literature has shown the role of IP6Ks in the central nervous system (CNS). IP6Ks have a key role in synaptic vesicle regulation, Akt/GSK3 signaling, neuronal migration, cell death, autophagy, nuclear translocation, and phosphate homeostasis. IP6Ks' regulation of these cellular processes has functional implications in vivo in behavior and CNS anatomy.
Collapse
Affiliation(s)
- Tyler Heitmann
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, 725 North Wolfe Street Suite 300, Baltimore, MD 21205, USA
- The Lieber Institute for Brain Development, 855 North Wolfe Street Suite 300, Baltimore, MD 21205, USA
| | - James C. Barrow
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, 725 North Wolfe Street Suite 300, Baltimore, MD 21205, USA
- The Lieber Institute for Brain Development, 855 North Wolfe Street Suite 300, Baltimore, MD 21205, USA
| |
Collapse
|
10
|
Xuan Z, Yang S, Clark B, Hill SE, Manning L, Colón-Ramos DA. The active zone protein Clarinet regulates synaptic sorting of ATG-9 and presynaptic autophagy. PLoS Biol 2023; 21:e3002030. [PMID: 37053235 PMCID: PMC10101500 DOI: 10.1371/journal.pbio.3002030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/08/2023] [Indexed: 04/14/2023] Open
Abstract
Autophagy is essential for cellular homeostasis and function. In neurons, autophagosome biogenesis is temporally and spatially regulated to occur near presynaptic sites, in part via the trafficking of autophagy transmembrane protein ATG-9. The molecules that regulate autophagy by sorting ATG-9 at synapses remain largely unknown. Here, we conduct forward genetic screens at single synapses of C. elegans neurons and identify a role for the long isoform of the active zone protein Clarinet (CLA-1L) in regulating sorting of autophagy protein ATG-9 at synapses, and presynaptic autophagy. We determine that disrupting CLA-1L results in abnormal accumulation of ATG-9 containing vesicles enriched with clathrin. The ATG-9 phenotype in cla-1(L) mutants is not observed for other synaptic vesicle proteins, suggesting distinct mechanisms that regulate sorting of ATG-9-containing vesicles and synaptic vesicles. Through genetic analyses, we uncover the adaptor protein complexes that genetically interact with CLA-1 in ATG-9 sorting. We also determine that CLA-1L extends from the active zone to the periactive zone and genetically interacts with periactive zone proteins in ATG-9 sorting. Our findings reveal novel roles for active zone proteins in the sorting of ATG-9 and in presynaptic autophagy.
Collapse
Affiliation(s)
- Zhao Xuan
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Departments of Neuroscience and Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Sisi Yang
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Departments of Neuroscience and Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Benjamin Clark
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Departments of Neuroscience and Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Sarah E. Hill
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Departments of Neuroscience and Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Laura Manning
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Departments of Neuroscience and Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Daniel A. Colón-Ramos
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Departments of Neuroscience and Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Instituto de Neurobiología, Recinto de Ciencias Médicas, Universidad de Puerto Rico, San Juan,Puerto Rico
- Wu Tsai Institute, Yale University, New Haven, Connecticut, United States of America
| |
Collapse
|
11
|
Wu XS, Wu LG. Multiple Modes of Fusion and Retrieval at the Calyx of Held Synapse. ADVANCES IN NEUROBIOLOGY 2023; 33:43-62. [PMID: 37615863 DOI: 10.1007/978-3-031-34229-5_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Neurotransmitter in vesicles is released through a fusion pore when vesicles fuse with the plasma membrane. Subsequent retrieval of the fused vesicle membrane is the key step in recycling exocytosed vesicles. Application of advanced electrophysiological techniques to a large nerve terminal, the calyx of Held, has led to recordings of endocytosis, individual vesicle fusion and retrieval, and the kinetics of the fusion pore opening process and the fission pore closure process. These studies have revealed three kinetically different forms of endocytosis-rapid, slow, and bulk-and two forms of fusion-full collapse and kiss-and-run. Calcium influx triggers all kinetically distinguishable forms of endocytosis at calyces by activation of calmodulin/calcineurin signaling pathway and protein kinase C, which may dephosphorylate and phosphorylate endocytic proteins. Polymerized actin may provide mechanical forces to bend the membrane, forming membrane pits, the precursor for generating vesicles. These research advancements are reviewed in this chapter.
Collapse
Affiliation(s)
- Xin-Sheng Wu
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA.
| | - Ling-Gang Wu
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA.
| |
Collapse
|
12
|
Illuminating membrane structural dynamics of fusion and endocytosis with advanced light imaging techniques. Biochem Soc Trans 2022; 50:1157-1167. [PMID: 35960003 PMCID: PMC9444071 DOI: 10.1042/bst20210263] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/24/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022]
Abstract
Visualization of cellular dynamics using fluorescent light microscopy has become a reliable and indispensable source of experimental evidence for biological studies. Over the past two decades, the development of super-resolution microscopy platforms coupled with innovations in protein and molecule labeling led to significant biological findings that were previously unobservable due to the barrier of the diffraction limit. As a result, the ability to image the dynamics of cellular processes is vastly enhanced. These imaging tools are extremely useful in cellular physiology for the study of vesicle fusion and endocytosis. In this review, we will explore the power of stimulated emission depletion (STED) and confocal microscopy in combination with various labeling techniques in real-time observation of the membrane transformation of fusion and endocytosis, as well as their underlying mechanisms. We will review how STED and confocal imaging are used to reveal fusion and endocytic membrane transformation processes in live cells, including hemi-fusion; hemi-fission; hemi-to-full fusion; fusion pore opening, expansion, constriction and closure; shrinking or enlargement of the Ω-shape membrane structure after vesicle fusion; sequential compound fusion; and the sequential endocytic membrane transformation from flat- to O-shape via the intermediate Λ- and Ω-shape transition. We will also discuss how the recent development of imaging techniques would impact future studies in the field.
Collapse
|
13
|
Li H, Datunashvili M, Reyes RC, Voglmaier SM. Inositol hexakisphosphate kinases differentially regulate trafficking of vesicular glutamate transporters 1 and 2. Front Cell Neurosci 2022; 16:926794. [PMID: 35936490 PMCID: PMC9355605 DOI: 10.3389/fncel.2022.926794] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/27/2022] [Indexed: 11/16/2022] Open
Abstract
Inositol pyrophosphates have been implicated in cellular signaling and membrane trafficking, including synaptic vesicle (SV) recycling. Inositol hexakisphosphate kinases (IP6Ks) and their product, diphosphoinositol pentakisphosphate (PP-IP5 or IP7), directly and indirectly regulate proteins important in vesicle recycling by the activity-dependent bulk endocytosis pathway (ADBE). In the present study, we show that two isoforms, IP6K1 and IP6K3, are expressed in axons. The role of the kinases in SV recycling are investigated using pharmacologic inhibition, shRNA knockdown, and IP6K1 and IP6K3 knockout mice. Live-cell imaging experiments use optical reporters of SV recycling based on vesicular glutamate transporter isoforms, VGLUT1- and VGLUT2-pHluorins (pH), which recycle differently. VGLUT1-pH recycles by classical AP-2 dependent endocytosis under moderate stimulation conditions, while VGLUT2-pH recycles using AP-1 and AP-3 adaptor proteins as well. Using a short stimulus to release the readily releasable pool (RRP), we show that IP6K1 KO increases exocytosis of both VGLUT1-and VGLUT2-pH, while IP6K3 KO decreases the amount of both transporters in the RRP. In electrophysiological experiments we measure glutamate signaling with short stimuli and under the intense stimulation conditions that trigger bulk endocytosis. IP6K1 KO increases synaptic facilitation and IP6K3 KO decreases facilitation compared to wild type in CA1 hippocampal Schaffer collateral synapses. After intense stimulation, the rate of endocytosis of VGLUT2-pH, but not VGLUT1-pH, is increased by knockout, knockdown, and pharmacologic inhibition of IP6Ks. Thus IP6Ks differentially affect the endocytosis of two SV protein cargos that use different endocytic pathways. However, while IP6K1 KO and IP6K3 KO exert similar effects on endocytosis after stimulation, the isoforms exert different effects on exocytosis earlier in the stimulus and on the early phase of glutamate release. Taken together, the data indicate a role for IP6Ks both in exocytosis early in the stimulation period and in endocytosis, particularly under conditions that may utilize AP-1/3 adaptors.
Collapse
|
14
|
Shi B, Jin YH, Wu LG. Dynamin 1 controls vesicle size and endocytosis at hippocampal synapses. Cell Calcium 2022; 103:102564. [PMID: 35220002 PMCID: PMC9009158 DOI: 10.1016/j.ceca.2022.102564] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/27/2022] [Accepted: 02/15/2022] [Indexed: 11/16/2022]
Abstract
Following calcium-triggered vesicle exocytosis, endocytosis regenerates vesicles to maintain exocytosis and thus synaptic transmission, which underlies neuronal circuit activities. Although most molecules involved in endocytosis have been identified, it remains rather poorly understood how endocytic machinery regulates vesicle size. Vesicle size, together with the transmitter concentration inside the vesicle, determines the amount of transmitter the vesicle can release, the quantal size, that may control the strength of synaptic transmission. Here, we report that, surprisingly, knockout of the GTPase dynamin 1, the most abundant brain dynamin isoform known to catalyze fission of the membrane pit's neck (the last step of endocytosis), not only significantly slowed endocytosis but also increased the synaptic vesicle diameter by as much as ∼40-64% at cultured hippocampal synapses. Furthermore, dynamin 1 knockout increased the size of membrane pits, the precursor for endocytic vesicle formation. These results suggest an important function of dynamin other than its well-known fission function - control of vesicle size at the pit formation stage.
Collapse
Affiliation(s)
- Bo Shi
- National Institute of Neurological Disorders and Stroke, 35 Convent Dr., Bethesda, MD 20892 United States; Biological Sciences Graduate Program, College of Computer, Mathematical, and Natural Sciences, University of Maryland, College Park, MD 20740 United States
| | - Ying-Hui Jin
- National Institute of Neurological Disorders and Stroke, 35 Convent Dr., Bethesda, MD 20892 United States; Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou 510515, China
| | - Ling-Gang Wu
- National Institute of Neurological Disorders and Stroke, 35 Convent Dr., Bethesda, MD 20892 United States.
| |
Collapse
|
15
|
Arpino G, Somasundaram A, Shin W, Ge L, Villareal S, Chan CY, Ashery U, Shupliakov O, Taraska JW, Wu LG. Clathrin-mediated endocytosis cooperates with bulk endocytosis to generate vesicles. iScience 2022; 25:103809. [PMID: 35198874 PMCID: PMC8841809 DOI: 10.1016/j.isci.2022.103809] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/02/2021] [Accepted: 01/20/2022] [Indexed: 11/25/2022] Open
Abstract
Clathrin-mediated endocytosis, the most prominent endocytic mode, is thought to be generated primarily from relatively flat patches of the plasma membrane. By employing conventional and platinum replica electron microscopy and super-resolution STED microscopy in neuroendocrine chromaffin cells, we found that large Ω-shaped or dome-shaped plasma membrane invaginations, previously thought of as the precursor of bulk endocytosis, are primary sites for clathrin-coated pit generation after depolarization. Clathrin-coated pits are more densely packed at invaginations rather than flat membranes, suggesting that invaginations are preferred sites for clathrin-coated pit formation, likely because their positive curvature facilitates coated-pit formation. Thus, clathrin-mediated endocytosis closely collaborates with bulk endocytosis to enhance endocytic capacity in active secretory cells. This direct collaboration between two classically independent endocytic pathways is of broad importance given the central role of both clathrin-mediated endocytosis and bulk endocytosis in neurons, endocrine cells, immune cells, and many other cell types throughout the body.
Collapse
Affiliation(s)
- Gianvito Arpino
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | | | - Wonchul Shin
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Lihao Ge
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Seth Villareal
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Chung Yu Chan
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Uri Ashery
- Life Science Faculty, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Oleg Shupliakov
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Institute of Translational Biomedicine, St Petersburg State University, St Petersburg, Russia
| | | | - Ling-Gang Wu
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| |
Collapse
|
16
|
Xia X, Wang Y, Qin Y, Zhao S, Zheng JC. Exosome: A novel neurotransmission modulator or non-canonical neurotransmitter? Ageing Res Rev 2022; 74:101558. [PMID: 34990846 DOI: 10.1016/j.arr.2021.101558] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/13/2021] [Accepted: 12/30/2021] [Indexed: 02/08/2023]
Abstract
Neurotransmission is the electrical impulse-triggered propagation of signals between neurons or between neurons and other cell types such as skeletal muscle cells. Recent studies point out the involvement of exosomes, a type of small bilipid layer-enclosed extracellular vesicles, in regulating neurotransmission. Through horizontally transferring proteins, lipids, and nucleic acids, exosomes can modulate synaptic activities rapidly by controlling neurotransmitter release or progressively by regulating neural plasticity including synapse formation, neurite growth & removal, and axon guidance & elongation. In this review, we summarize the similarities and differences between exosomes and synaptic vesicles in their biogenesis, contents, and release. We also highlight the recent progress made in demonstrating the biological roles of exosome in regulating neurotransmission, and propose a modified model of neurotransmission, in which exosomes act as novel neurotransmitters. Lastly, we provide a comprehensive discussion of the enlightenment of the current knowledge on neurotransmission to the future directions of exosome research.
Collapse
|
17
|
López-Hernández T, Takenaka KI, Mori Y, Kongpracha P, Nagamori S, Haucke V, Takamori S. Clathrin-independent endocytic retrieval of SV proteins mediated by the clathrin adaptor AP-2 at mammalian central synapses. eLife 2022; 11:e71198. [PMID: 35014951 PMCID: PMC8752090 DOI: 10.7554/elife.71198] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 12/28/2021] [Indexed: 12/25/2022] Open
Abstract
Neurotransmission is based on the exocytic fusion of synaptic vesicles (SVs) followed by endocytic membrane retrieval and the reformation of SVs. Conflicting models have been proposed regarding the mechanisms of SV endocytosis, most notably clathrin/adaptor protein complex 2 (AP-2)-mediated endocytosis and clathrin-independent ultrafast endocytosis. Partitioning between these pathways has been suggested to be controlled by temperature and stimulus paradigm. We report on the comprehensive survey of six major SV proteins to show that SV endocytosis in mouse hippocampal neurons at physiological temperature occurs independent of clathrin while the endocytic retrieval of a subset of SV proteins including the vesicular transporters for glutamate and GABA depend on sorting by the clathrin adaptor AP-2. Our findings highlight a clathrin-independent role of the clathrin adaptor AP-2 in the endocytic retrieval of select SV cargos from the presynaptic cell surface and suggest a revised model for the endocytosis of SV membranes at mammalian central synapses.
Collapse
Affiliation(s)
| | - Koh-ichiro Takenaka
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha UniversityKyotoJapan
| | - Yasunori Mori
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha UniversityKyotoJapan
| | - Pornparn Kongpracha
- Department of Laboratory Medicine, The Jikei University School of MedicineTokyoJapan
| | - Shushi Nagamori
- Department of Laboratory Medicine, The Jikei University School of MedicineTokyoJapan
| | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP)BerlinGermany
| | - Shigeo Takamori
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha UniversityKyotoJapan
| |
Collapse
|
18
|
Peng YJ, Geng J, Wu Y, Pinales C, Langen J, Chang YC, Buser C, Chang KT. Minibrain kinase and calcineurin coordinate activity-dependent bulk endocytosis through synaptojanin. J Cell Biol 2021; 220:212674. [PMID: 34596663 PMCID: PMC8491876 DOI: 10.1083/jcb.202011028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 07/28/2021] [Accepted: 09/16/2021] [Indexed: 11/22/2022] Open
Abstract
Neurons use multiple modes of endocytosis, including clathrin-mediated endocytosis (CME) and activity-dependent bulk endocytosis (ADBE), during mild and intense neuronal activity, respectively, to maintain stable neurotransmission. While molecular players modulating CME are well characterized, factors regulating ADBE and mechanisms coordinating CME and ADBE activations remain poorly understood. Here we report that Minibrain/DYRK1A (Mnb), a kinase mutated in autism and up-regulated in Down's syndrome, plays a novel role in suppressing ADBE. We demonstrate that Mnb, together with calcineurin, delicately coordinates CME and ADBE by controlling the phosphoinositol phosphatase activity of synaptojanin (Synj) during varying synaptic demands. Functional domain analyses reveal that Synj's 5'-phosphoinositol phosphatase activity suppresses ADBE, while SAC1 activity is required for efficient ADBE. Consequently, Parkinson's disease mutation in Synj's SAC1 domain impairs ADBE. These data identify Mnb and Synj as novel regulators of ADBE and further indicate that CME and ADBE are differentially governed by Synj's dual phosphatase domains.
Collapse
Affiliation(s)
- Yi-Jheng Peng
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA.,Neuroscience Graduate Program, University of Southern California, Los Angeles, CA
| | - Junhua Geng
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Ying Wu
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | | | - Jennifer Langen
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Yen-Ching Chang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | | | - Karen T Chang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA.,Neuroscience Graduate Program, University of Southern California, Los Angeles, CA.,Department of Physiology & Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA
| |
Collapse
|
19
|
Sauvola CW, Littleton JT. SNARE Regulatory Proteins in Synaptic Vesicle Fusion and Recycling. Front Mol Neurosci 2021; 14:733138. [PMID: 34421538 PMCID: PMC8377282 DOI: 10.3389/fnmol.2021.733138] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 07/20/2021] [Indexed: 01/01/2023] Open
Abstract
Membrane fusion is a universal feature of eukaryotic protein trafficking and is mediated by the soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) family. SNARE proteins embedded in opposing membranes spontaneously assemble to drive membrane fusion and cargo exchange in vitro. Evolution has generated a diverse complement of SNARE regulatory proteins (SRPs) that ensure membrane fusion occurs at the right time and place in vivo. While a core set of SNAREs and SRPs are common to all eukaryotic cells, a specialized set of SRPs within neurons confer additional regulation to synaptic vesicle (SV) fusion. Neuronal communication is characterized by precise spatial and temporal control of SNARE dynamics within presynaptic subdomains specialized for neurotransmitter release. Action potential-elicited Ca2+ influx at these release sites triggers zippering of SNAREs embedded in the SV and plasma membrane to drive bilayer fusion and release of neurotransmitters that activate downstream targets. Here we discuss current models for how SRPs regulate SNARE dynamics and presynaptic output, emphasizing invertebrate genetic findings that advanced our understanding of SRP regulation of SV cycling.
Collapse
Affiliation(s)
- Chad W Sauvola
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - J Troy Littleton
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
20
|
Binotti B, Jahn R, Pérez-Lara Á. An overview of the synaptic vesicle lipid composition. Arch Biochem Biophys 2021; 709:108966. [PMID: 34139199 DOI: 10.1016/j.abb.2021.108966] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 06/10/2021] [Accepted: 06/10/2021] [Indexed: 11/29/2022]
Abstract
Chemical neurotransmission is the major mechanism of neuronal communication. Neurotransmitters are released from secretory organelles, the synaptic vesicles (SVs) via exocytosis into the synaptic cleft. Fusion of SVs with the presynaptic plasma membrane is balanced by endocytosis, thus maintaining the presynaptic membrane at steady-state levels. The protein machineries responsible for exo- and endocytosis have been extensively investigated. In contrast, less is known about the role of lipids in synaptic transmission and how the lipid composition of SVs is affected by dynamic exo-endocytotic cycling. Here we summarize the current knowledge about the composition, organization, and function of SV membrane lipids. We also cover lipid biogenesis and maintenance during the synaptic vesicle cycle.
Collapse
Affiliation(s)
- Beyenech Binotti
- Department of Biochemistry, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Reinhard Jahn
- Department of Neurobiology, Max-Planck-Institute for Biophysical Chemistry, Am Faßberg 11, 37077, Göttingen, Germany.
| | - Ángel Pérez-Lara
- Department of Physical Chemistry, University of Granada, Campus Universitario de Cartuja, 18071, Granada, Spain.
| |
Collapse
|
21
|
Camblor-Perujo S, Kononenko NL. Brain-specific functions of the endocytic machinery. FEBS J 2021; 289:2219-2246. [PMID: 33896112 DOI: 10.1111/febs.15897] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/29/2021] [Indexed: 12/12/2022]
Abstract
Endocytosis is an essential cellular process required for multiple physiological functions, including communication with the extracellular environment, nutrient uptake, and signaling by the cell surface receptors. In a broad sense, endocytosis is accomplished through either constitutive or ligand-induced invagination of the plasma membrane, which results in the formation of the plasma membrane-retrieved endocytic vesicles, which can either be sent for degradation to the lysosomes or recycled back to the PM. This additional function of endocytosis in membrane retrieval has been adopted by excitable cells, such as neurons, for membrane equilibrium maintenance at synapses. The last two decades were especially productive with respect to the identification of brain-specific functions of the endocytic machinery, which additionally include but not limited to regulation of neuronal differentiation and migration, maintenance of neuron morphology and synaptic plasticity, and prevention of neurotoxic aggregates spreading. In this review, we highlight the current knowledge of brain-specific functions of endocytic machinery with a specific focus on three brain cell types, neuronal progenitor cells, neurons, and glial cells.
Collapse
Affiliation(s)
| | - Natalia L Kononenko
- CECAD Cluster of Excellence, University of Cologne, Germany.,Center for Physiology & Pathophysiology, Medical Faculty, University of Cologne, Germany
| |
Collapse
|
22
|
Recruitment of Polarity Complexes and Tight Junction Proteins to the Site of Apical Bulk Endocytosis. Cell Mol Gastroenterol Hepatol 2021; 12:59-80. [PMID: 33548596 PMCID: PMC8082271 DOI: 10.1016/j.jcmgh.2021.01.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS The molecular motor, Myosin Vb (MYO5B), is well documented for its role in trafficking cargo to the apical membrane of epithelial cells. Despite its involvement in regulating apical proteins, the role of MYO5B in cell polarity is less clear. Inactivating mutations in MYO5B result in microvillus inclusion disease (MVID), a disorder characterized by loss of key apical transporters and the presence of intracellular inclusions in enterocytes. We previously identified that inclusions in Myo5b knockout (KO) mice form from invagination of the apical brush border via apical bulk endocytosis. Herein, we sought to elucidate the role of polarity complexes and tight junction proteins during the formation of inclusions. METHODS Intestinal tissue from neonatal control and Myo5b KO littermates was analyzed by immunofluorescence to determine the localization of polarity complexes and tight junction proteins. RESULTS Proteins that make up the apical polarity complexes-Crumbs3 and Pars complexes-were associated with inclusions in Myo5b KO mice. In addition, tight junction proteins were observed to be concentrated over inclusions that were present at the apical membrane of Myo5b-deficient enterocytes in vivo and in vitro. Our mouse findings are complemented by immunostaining in a large animal swine model of MVID genetically engineered to express a human MVID-associated mutation that shows an accumulation of Claudin-2 over forming inclusions. The findings from our swine model of MVID suggest that a similar mechanism of tight junction accumulation occurs in patients with MVID. CONCLUSIONS These data show that apical bulk endocytosis involves the altered localization of apical polarity proteins and tight junction proteins after loss of Myo5b.
Collapse
|
23
|
Winner BM, Bodt SML, McNutt PM. Special Delivery: Potential Mechanisms of Botulinum Neurotoxin Uptake and Trafficking within Motor Nerve Terminals. Int J Mol Sci 2020; 21:ijms21228715. [PMID: 33218099 PMCID: PMC7698961 DOI: 10.3390/ijms21228715] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/11/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
Botulinum neurotoxins (BoNTs) are highly potent, neuroparalytic protein toxins that block the release of acetylcholine from motor neurons and autonomic synapses. The unparalleled toxicity of BoNTs results from the highly specific and localized cleavage of presynaptic proteins required for nerve transmission. Currently, the only pharmacotherapy for botulism is prophylaxis with antitoxin, which becomes progressively less effective as symptoms develop. Treatment for symptomatic botulism is limited to supportive care and artificial ventilation until respiratory function spontaneously recovers, which can take weeks or longer. Mechanistic insights into intracellular toxin behavior have progressed significantly since it was shown that toxins exploit synaptic endocytosis for entry into the nerve terminal, but fundamental questions about host-toxin interactions remain unanswered. Chief among these are mechanisms by which BoNT is internalized into neurons and trafficked to sites of molecular toxicity. Elucidating how receptor-bound toxin is internalized and conditions under which the toxin light chain engages with target SNARE proteins is critical for understanding the dynamics of intoxication and identifying novel therapeutics. Here, we discuss the implications of newly discovered modes of synaptic vesicle recycling on BoNT uptake and intraneuronal trafficking.
Collapse
Affiliation(s)
- Brittany M. Winner
- United States Army Medical Research Institute of Chemical Defense, Gunpowder, MD 21047, USA;
| | - Skylar M. L. Bodt
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033, USA;
| | - Patrick M. McNutt
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston-Salem, NC 27101, USA
- Correspondence:
| |
Collapse
|
24
|
Overhoff M, De Bruyckere E, Kononenko NL. Mechanisms of neuronal survival safeguarded by endocytosis and autophagy. J Neurochem 2020; 157:263-296. [PMID: 32964462 DOI: 10.1111/jnc.15194] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/21/2020] [Accepted: 09/08/2020] [Indexed: 12/11/2022]
Abstract
Multiple aspects of neuronal physiology crucially depend on two cellular pathways, autophagy and endocytosis. During endocytosis, extracellular components either unbound or recognized by membrane-localized receptors (termed "cargo") become internalized into plasma membrane-derived vesicles. These can serve to either recycle the material back to the plasma membrane or send it for degradation to lysosomes. Autophagy also uses lysosomes as a terminal degradation point, although instead of degrading the plasma membrane-derived cargo, autophagy eliminates detrimental cytosolic material and intracellular organelles, which are transported to lysosomes by means of double-membrane vesicles, referred to as autophagosomes. Neurons, like all non-neuronal cells, capitalize on autophagy and endocytosis to communicate with the environment and maintain protein and organelle homeostasis. Additionally, the highly polarized, post-mitotic nature of neurons made them adopt these two pathways for cell-specific functions. These include the maintenance of the synaptic vesicle pool in the pre-synaptic terminal and the long-distance transport of signaling molecules. Originally discovered independently from each other, it is now clear that autophagy and endocytosis are closely interconnected and share several common participating molecules. Considering the crucial role of autophagy and endocytosis in cell type-specific functions in neurons, it is not surprising that defects in both pathways have been linked to the pathology of numerous neurodegenerative diseases. In this review, we highlight the recent knowledge of the role of endocytosis and autophagy in neurons with a special focus on synaptic physiology and discuss how impairments in genes coding for autophagy and endocytosis proteins can cause neurodegeneration.
Collapse
Affiliation(s)
- Melina Overhoff
- CECAD Cluster of Excellence, Institute for Genetics, University of Cologne, Cologne, Germany
| | - Elodie De Bruyckere
- CECAD Cluster of Excellence, Institute for Genetics, University of Cologne, Cologne, Germany
| | - Natalia L Kononenko
- CECAD Cluster of Excellence, Institute for Genetics, University of Cologne, Cologne, Germany
| |
Collapse
|
25
|
Tao-Cheng JH. Stimulation-induced differential redistributions of clathrin and clathrin-coated vesicles in axons compared to soma/dendrites. Mol Brain 2020; 13:141. [PMID: 33066817 PMCID: PMC7565815 DOI: 10.1186/s13041-020-00683-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/08/2020] [Indexed: 11/17/2022] Open
Abstract
Clathrin-mediated endocytosis plays an important role in the recycling of synaptic vesicle in presynaptic terminals, and in the recycling of transmitter receptors in neuronal soma/dendrites. The present study uses electron microscopy (EM) and immunogold EM to document the different categories of clathrin-coated vesicles (CCV) and pits (CCP) in axons compared to soma/dendrites, and the depolarization-induced redistribution of clathrin in these two polarized compartments of the neuron. The size of CCVs in presynaptic terminals (~ 40 nm; similar to the size of synaptic vesicles) is considerably smaller than the size of CCVs in soma/dendrites (~ 90 nm). Furthermore, neuronal stimulation induces an increase in the number of CCV/CCP in presynaptic terminals, but a decrease in soma/dendrites. Immunogold labeling of clathrin revealed that in presynaptic terminals under resting conditions, the majority of clathrin molecules are unassembled and concentrated outside of synaptic vesicle clusters. Upon depolarization with high K+, label for clathrin became scattered among de-clustered synaptic vesicles and moved closer to the presynaptic active zone. In contrast to axons, clathrin-labeled CCVs and CCPs were prominent in soma/dendrites under resting conditions, and became inconspicuous upon depolarization with high K+. Thus, EM examination suggests that the regulation and mechanism of clathrin-mediated endocytosis differ between axon and dendrite, and that clathrin redistributes differently in these two neuronal compartments upon depolarization.
Collapse
Affiliation(s)
- Jung-Hwa Tao-Cheng
- NINDS Electron Microscopy Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
26
|
d'Errico P, Meyer-Luehmann M. Mechanisms of Pathogenic Tau and Aβ Protein Spreading in Alzheimer's Disease. Front Aging Neurosci 2020; 12:265. [PMID: 33061903 PMCID: PMC7481386 DOI: 10.3389/fnagi.2020.00265] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/03/2020] [Indexed: 01/01/2023] Open
Abstract
Alzheimer’s disease (AD) is pathologically defined by extracellular accumulation of amyloid-β (Aβ) peptides generated by the cleavage of amyloid precursor protein (APP), strings of hyperphosphorylated Tau proteins accumulating inside neurons known as neurofibrillary tangles (NFTs) and neuronal loss. The association between the two hallmarks and cognitive decline has been known since the beginning of the 20th century when the first description of the disease was carried out by Alois Alzheimer. Today, more than 40 million people worldwide are affected by AD that represents the most common cause of dementia and there is still no effective treatment available to cure the disease. In general, the aggregation of Aβ is considered an essential trigger in AD pathogenesis that gives rise to NFTs, neuronal dysfunction and dementia. During the process leading to AD, tau and Aβ first misfold and form aggregates in one brain region, from where they spread to interconnected areas of the brain thereby inducing its gradual morphological and functional deterioration. In this mini-review article, we present an overview of the current literature on the spreading mechanisms of Aβ and tau pathology in AD since a more profound understanding is necessary to design therapeutic approaches aimed at preventing or halting disease progression.
Collapse
Affiliation(s)
- Paolo d'Errico
- Department of Neurology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Meyer-Luehmann
- Department of Neurology, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
27
|
Gowrisankaran S, Houy S, Del Castillo JGP, Steubler V, Gelker M, Kroll J, Pinheiro PS, Schwitters D, Halbsgut N, Pechstein A, van Weering JRT, Maritzen T, Haucke V, Raimundo N, Sørensen JB, Milosevic I. Endophilin-A coordinates priming and fusion of neurosecretory vesicles via intersectin. Nat Commun 2020; 11:1266. [PMID: 32152276 PMCID: PMC7062783 DOI: 10.1038/s41467-020-14993-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 02/12/2020] [Indexed: 12/17/2022] Open
Abstract
Endophilins-A are conserved endocytic adaptors with membrane curvature-sensing and -inducing properties. We show here that, independently of their role in endocytosis, endophilin-A1 and endophilin-A2 regulate exocytosis of neurosecretory vesicles. The number and distribution of neurosecretory vesicles were not changed in chromaffin cells lacking endophilin-A, yet fast capacitance and amperometry measurements revealed reduced exocytosis, smaller vesicle pools and altered fusion kinetics. The levels and distributions of the main exocytic and endocytic factors were unchanged, and slow compensatory endocytosis was not robustly affected. Endophilin-A’s role in exocytosis is mediated through its SH3-domain, specifically via a direct interaction with intersectin-1, a coordinator of exocytic and endocytic traffic. Endophilin-A not able to bind intersectin-1, and intersectin-1 not able to bind endophilin-A, resulted in similar exocytic defects in chromaffin cells. Altogether, we report that two endocytic proteins, endophilin-A and intersectin-1, are enriched on neurosecretory vesicles and regulate exocytosis by coordinating neurosecretory vesicle priming and fusion. Endophilins-A are conserved membrane-associated proteins required for endocytosis. Here, the authors report that endophilins-A also promote exocytosis of neurosecretory vesicles by coordinating priming and fusion through intersectin-1, independently of their roles in different types of endocytosis.
Collapse
Affiliation(s)
- Sindhuja Gowrisankaran
- European Neuroscience Institute-A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society Göttingen, Göttingen, Germany
| | - Sébastien Houy
- University of Copenhagen, Department for Neuroscience, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Johanna G Peña Del Castillo
- European Neuroscience Institute-A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society Göttingen, Göttingen, Germany
| | - Vicky Steubler
- European Neuroscience Institute-A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society Göttingen, Göttingen, Germany
| | - Monika Gelker
- European Neuroscience Institute-A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society Göttingen, Göttingen, Germany
| | - Jana Kroll
- European Neuroscience Institute-A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society Göttingen, Göttingen, Germany
| | - Paulo S Pinheiro
- University of Copenhagen, Department for Neuroscience, Faculty of Health and Medical Sciences, Copenhagen, Denmark.,Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Dirk Schwitters
- European Neuroscience Institute-A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society Göttingen, Göttingen, Germany
| | - Nils Halbsgut
- European Neuroscience Institute-A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society Göttingen, Göttingen, Germany
| | - Arndt Pechstein
- Leibniz Research Institute for Molecular Pharmacology, Molecular Physiology and Cell Biology Section, Berlin, Germany
| | - Jan R T van Weering
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam UMC, Amsterdam, The Netherlands
| | - Tanja Maritzen
- Leibniz Research Institute for Molecular Pharmacology, Molecular Physiology and Cell Biology Section, Berlin, Germany
| | - Volker Haucke
- Leibniz Research Institute for Molecular Pharmacology, Molecular Physiology and Cell Biology Section, Berlin, Germany
| | - Nuno Raimundo
- Institute for Cellular Biochemistry, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Jakob B Sørensen
- University of Copenhagen, Department for Neuroscience, Faculty of Health and Medical Sciences, Copenhagen, Denmark.
| | - Ira Milosevic
- European Neuroscience Institute-A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society Göttingen, Göttingen, Germany.
| |
Collapse
|
28
|
Venkatesh K, Mathew A, Koushika SP. Role of actin in organelle trafficking in neurons. Cytoskeleton (Hoboken) 2020; 77:97-109. [DOI: 10.1002/cm.21580] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/14/2019] [Accepted: 11/05/2019] [Indexed: 01/29/2023]
Affiliation(s)
- Keertana Venkatesh
- Department of Biological SciencesTata Institute of Fundamental Research Mumbai India
| | - Amal Mathew
- Department of Biological SciencesTata Institute of Fundamental Research Mumbai India
| | - Sandhya P. Koushika
- Department of Biological SciencesTata Institute of Fundamental Research Mumbai India
| |
Collapse
|
29
|
Bilkey J, Nahirney PC, Delaney KR. Time and exposure to serotonin affect releasability of recycled vesicles at crayfish claw opener muscle synapses. Synapse 2019; 74:e22136. [PMID: 31574172 DOI: 10.1002/syn.22136] [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: 08/09/2019] [Revised: 09/16/2019] [Accepted: 09/21/2019] [Indexed: 11/10/2022]
Abstract
The crayfish claw opener neuromuscular junction is a biological model for studying presynaptic neuromodulation by serotonin (5HT) and synaptic vesicle recycling. It has been hypothesized that 5HT enhances release by recruiting a population of either previously nonrecycling or "reluctant" vesicles to increase the readily releasable pool. To determine if 5HT activates a distinct population of synaptic vesicles, recycling membranes were labeled with the membrane dye, FM1-43. Unloading (destaining) protocols could not resolve a population of vesicles that were only releasable in the presence of 5HT. Instead, we conclude synaptic vesicles change behavior in axon terminals independent of 5HT, becoming less likely to exocytose and unload dye over periods of >1 hr after recycling. We hypothesized this to be due to the slow conversion of a portion of recycled vesicles to a difficult to release state. The possibility that vesicles in these pools were spatially separated within the terminal was tested using photoconversion of FM1-43 and transmission electron microscopy. The location of FM1-43-labeled vesicles fixed 2 min following 3 min of 20-Hz stimulation did not reveal preferential localization of recycling vesicles specifically near release sites and the distribution of labeled vesicles was not significantly different between early (2 min) and late (180 min) time points. Terminals fixed 30 s following stimulation contained a significant proportion of vesicular structures equivalent in diameter to 2-5 regular vesicles, with multivesicular bodies and calveoli rarely seen, suggesting that endocytosis during sustained release at crayfish terminals occurs via multiple routes, most commonly through large "vesicle" intermediates.
Collapse
Affiliation(s)
- Jessica Bilkey
- Department of Biology, University of Victoria, Victoria, British Columbia, Canada
| | - Patrick C Nahirney
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Kerry R Delaney
- Department of Biology, University of Victoria, Victoria, British Columbia, Canada
| |
Collapse
|
30
|
Engevik AC, Kaji I, Postema MM, Faust JJ, Meyer AR, Williams JA, Fitz GN, Tyska MJ, Wilson JM, Goldenring JR. Loss of myosin Vb promotes apical bulk endocytosis in neonatal enterocytes. J Cell Biol 2019; 218:3647-3662. [PMID: 31562230 PMCID: PMC6829668 DOI: 10.1083/jcb.201902063] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/22/2019] [Accepted: 08/29/2019] [Indexed: 12/22/2022] Open
Abstract
In patients with inactivating mutations in myosin Vb (Myo5B), enterocytes show large inclusions lined by microvilli. The origin of inclusions in small-intestinal enterocytes in microvillus inclusion disease is currently unclear. We postulated that inclusions in Myo5b KO mouse enterocytes form through invagination of the apical brush border membrane. 70-kD FITC-dextran added apically to Myo5b KO intestinal explants accumulated in intracellular inclusions. Live imaging of Myo5b KO-derived enteroids confirmed the formation of inclusions from the apical membrane. Treatment of intestinal explants and enteroids with Dyngo resulted in accumulation of inclusions at the apical membrane. Inclusions in Myo5b KO enterocytes contained VAMP4 and Pacsin 2 (Syndapin 2). Myo5b;Pacsin 2 double-KO mice showed a significant decrease in inclusion formation. Our results suggest that apical bulk endocytosis in Myo5b KO enterocytes resembles activity-dependent bulk endocytosis, the primary mechanism for synaptic vesicle uptake during intense neuronal stimulation. Thus, apical bulk endocytosis mediates the formation of inclusions in neonatal Myo5b KO enterocytes.
Collapse
Affiliation(s)
- Amy C Engevik
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN.,Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Izumi Kaji
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN.,Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Meagan M Postema
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - James J Faust
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Anne R Meyer
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Janice A Williams
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN.,The Epithelial Biology Center and Vanderbilt University School of Medicine, Nashville, TN
| | - Gillian N Fitz
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN.,The Epithelial Biology Center and Vanderbilt University School of Medicine, Nashville, TN
| | - Jean M Wilson
- Department of Cellular and Molecular Medicine, Bio5 Institute, University of Arizona, Tucson, AZ
| | - James R Goldenring
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN .,Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN.,The Epithelial Biology Center and Vanderbilt University School of Medicine, Nashville, TN.,The Nashville VA Medical Center, Nashville, TN
| |
Collapse
|
31
|
Watanabe S, Mamer LE, Raychaudhuri S, Luvsanjav D, Eisen J, Trimbuch T, Söhl-Kielczynski B, Fenske P, Milosevic I, Rosenmund C, Jorgensen EM. Synaptojanin and Endophilin Mediate Neck Formation during Ultrafast Endocytosis. Neuron 2019; 98:1184-1197.e6. [PMID: 29953872 DOI: 10.1016/j.neuron.2018.06.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 05/12/2018] [Accepted: 06/04/2018] [Indexed: 11/19/2022]
Abstract
Ultrafast endocytosis generates vesicles from the plasma membrane as quickly as 50 ms in hippocampal neurons following synaptic vesicle fusion. The molecular mechanism underlying the rapid maturation of these endocytic pits is not known. Here we demonstrate that synaptojanin-1, and its partner endophilin-A, function in ultrafast endocytosis. In the absence of synaptojanin or endophilin, the membrane is rapidly invaginated, but pits do not become constricted at the base. The 5-phosphatase activity of synaptojanin is involved in formation of the neck, but 4-phosphatase is not required. Nevertheless, these pits are eventually cleaved into vesicles; within a 30-s interval, synaptic endosomes form and are resolved by clathrin-mediated budding. Then synaptojanin and endophilin function at a second step to aid with the removal of clathrin coats from the regenerated vesicles. These data together suggest that synaptojanin and endophilin can mediate membrane remodeling on a millisecond timescale during ultrafast endocytosis.
Collapse
Affiliation(s)
- Shigeki Watanabe
- Department of Neurophysiology, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany; Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA.
| | - Lauren Elizabeth Mamer
- Department of Neurophysiology, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany; The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Sumana Raychaudhuri
- Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Delgermaa Luvsanjav
- Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Julia Eisen
- Barnard College of Columbia University, New York, NY, USA
| | - Thorsten Trimbuch
- Department of Neurophysiology, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Berit Söhl-Kielczynski
- Department of Neurophysiology, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Pascal Fenske
- Department of Neurophysiology, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ira Milosevic
- Synaptic Vesicle Dynamics, European Neuroscience Institute, University Medical Center Göttingen, 37077 Göttingen, Germany
| | - Christian Rosenmund
- Department of Neurophysiology, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Erik M Jorgensen
- Department of Neurophysiology, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany; Department of Biology and Howard Hughes Medical Institute, University of Utah, Salt Lake City, UT 84112-0840, USA.
| |
Collapse
|
32
|
Saldeitis K, Richter K, Fischer KD, Ohl FW, Mateos JM, Budinger E. Ultrastructure of giant thalamic terminals in the auditory cortex. Eur J Neurosci 2019; 50:3445-3453. [PMID: 31286598 DOI: 10.1111/ejn.14509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 06/24/2019] [Accepted: 06/27/2019] [Indexed: 11/28/2022]
Abstract
The auditory system comprises some very large axonal terminals like the endbulb and calyx of Held and "giant" corticothalamic synapses. Previously, we described a hitherto unknown population of giant thalamocortical boutons arising from the medial division of the medial geniculate body (MGm) in the Mongolian gerbil, which terminate over a wide cortical range but in a columnar manner particularly in the extragranular layers of the auditory cortex. As a first step towards an understanding of their potential functional role, we here describe their ultrastructure combining anterograde tract-tracing with biocytin and electron microscopy. Quantitative ultrastructural analyses revealed that biocytin-labelled MGm boutons reach much larger sizes than other, non-labelled boutons. Also, mitochondria occupy more space within labelled boutons whereas synapses are of similar size. Labelled boutons are very heterogeneous in size but homogeneous with respect to their ultrastructural characteristics, with asymmetric synapses containing clear, round vesicles and targeting dendritic spines. Functionally, the ultrastructure of the MGm terminals indicates that they form excitatory contacts, which may transmit their information in a rapid, powerful and high-fidelity manner onto strategically advantageous compartments of their cortical target cells.
Collapse
Affiliation(s)
- Katja Saldeitis
- Department Systems Physiology of Learning, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Cognitive Hearing in Primates Group, Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany
| | - Karin Richter
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Klaus-Dieter Fischer
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Frank W Ohl
- Department Systems Physiology of Learning, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - José M Mateos
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Eike Budinger
- Department Systems Physiology of Learning, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany
| |
Collapse
|
33
|
Jin Y, Seo KH, Ko HM, Jung TW, Chung YH, Lee JH, Park HH, Kim HC, Jeong JH, Lee SH. Various approaches for measurement of synaptic vesicle endocytosis at the central nerve terminal. Arch Pharm Res 2019; 42:455-465. [PMID: 31115782 DOI: 10.1007/s12272-019-01161-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 05/16/2019] [Indexed: 10/26/2022]
Abstract
At the presynaptic terminal, neurotransmitters are stored in synaptic vesicles (SVs), which are released and recycled via exo- and endocytosis. SV endocytosis is crucial for sustaining synaptic transmission by maintaining the SV pool. Many studies have shown that presynaptic dysfunction, particularly impairment of SV endocytosis, is related to neurological disorders. Notably, the presynaptic terminal is considered to be a sensitive structure because certain presynaptic dysfunctions, manifested as impaired SV endocytosis or ultrastructural changes in the presynaptic terminal, can be observed before there is a biochemical or pathological evidence of a neurological disorder. Therefore, monitoring and assessing the presynaptic function by SV endocytosis facilitates the development of early markers for neurological disorders. In this study, we reviewed the current methods for assessing and visualizing SV endocytosis at the central nerve terminal.
Collapse
Affiliation(s)
- Yeonsun Jin
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Kyoung Hee Seo
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Hyun Myung Ko
- Department of Life Science, College of Science and Technology, Woosuk University, Jincheon, 27841, Republic of Korea
| | - Tae Woo Jung
- Research Administration Team, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Yoon Hee Chung
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Jong Hyuk Lee
- Department of Pharmaceutical Engineering, College of Life and Health Science, Hoseo University, Asan, 31499, Republic of Korea
| | - Hyun Ho Park
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Sung Hoon Lee
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
34
|
Yu A, Fox SG, Cavallini A, Kerridge C, O'Neill MJ, Wolak J, Bose S, Morimoto RI. Tau protein aggregates inhibit the protein-folding and vesicular trafficking arms of the cellular proteostasis network. J Biol Chem 2019; 294:7917-7930. [PMID: 30936201 DOI: 10.1074/jbc.ra119.007527] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/29/2019] [Indexed: 11/06/2022] Open
Abstract
Tauopathies are a diverse class of neurodegenerative diseases characterized by the formation of insoluble tau aggregates and the loss of cellular function and neuronal death. Tau inclusions have been shown to contain a number of proteins, including molecular chaperones, but the consequences of these entrapments are not well established. Here, using a human cell system for seeding-dependent tau aggregation, we demonstrate that the molecular chaperones heat-shock cognate 71-kDa protein (HSC70)/heat-shock protein 70 (HSP70), HSP90, and J-domain co-chaperones are sequestered by tau aggregates. By employing single-cell analysis of protein-folding and clathrin-mediated endocytosis, we show that both chaperone-dependent cellular activities are significantly impaired by tau aggregation and can be reversed by treatment with small-molecule regulators of heat-shock transcription factor 1 (HSF1) proteostasis that induce the expression of cytosolic chaperones. These results reveal that the sequestration of cytoplasmic molecular chaperones by tau aggregates interferes with two arms of the proteostasis network, likely having profound negative consequences for cellular function.
Collapse
Affiliation(s)
- Anan Yu
- From the Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208 and
| | - Susan G Fox
- From the Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208 and
| | - Annalisa Cavallini
- the Lilly Research Centre, Eli Lilly and Co. Ltd., Erl Wood Manor, Sunninghill Road, Windlesham, Surrey GU20 6PH, United Kingdom
| | - Caroline Kerridge
- the Lilly Research Centre, Eli Lilly and Co. Ltd., Erl Wood Manor, Sunninghill Road, Windlesham, Surrey GU20 6PH, United Kingdom
| | - Michael J O'Neill
- the Lilly Research Centre, Eli Lilly and Co. Ltd., Erl Wood Manor, Sunninghill Road, Windlesham, Surrey GU20 6PH, United Kingdom
| | - Joanna Wolak
- the Lilly Research Centre, Eli Lilly and Co. Ltd., Erl Wood Manor, Sunninghill Road, Windlesham, Surrey GU20 6PH, United Kingdom
| | - Suchira Bose
- the Lilly Research Centre, Eli Lilly and Co. Ltd., Erl Wood Manor, Sunninghill Road, Windlesham, Surrey GU20 6PH, United Kingdom
| | - Richard I Morimoto
- From the Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208 and
| |
Collapse
|
35
|
Kinetic Measurements of Endocytosis and Exocytosis in Cultured Neurons. Methods Mol Biol 2019. [PMID: 30771167 DOI: 10.1007/978-1-4939-9124-2_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The measurement of synaptic vesicle recycling in live neurons transfected with vesicular glutamate transporter fused to pHluorin (vGLUT-pHluorin) allows us to study exocytosis and endocytosis in neurons. When neurons are transfected with this protein we can measure the rate of vesicles fusing and internalizing from the membrane using live total internal reflection fluorescence (TIRF) imaging. Here, we describe transfection, culturing, and imaging of wild-type and αβγ-synuclein knockout hippocampal neurons. This technique can be used to evaluate the effect of different phenotypes and treatments in the physiology of synaptic vesicle recycling in cultured neurons.
Collapse
|
36
|
Extracting lipid vesicles from plasma membranes via self-assembly of clathrin-inspired scaffolding nanoparticles. Colloids Surf B Biointerfaces 2019; 176:239-248. [PMID: 30623811 DOI: 10.1016/j.colsurfb.2019.01.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/27/2018] [Accepted: 01/02/2019] [Indexed: 11/22/2022]
Abstract
Single-cell analysis is a new and rapidly expanding field, the goal of which is obtaining fresh information from individual cells to understand the regulatory mechanisms of cell development and diseases. Conventional approaches generally rely on the cell lysis which, however, is destructive to cells and against multiple sampling from the living cell. Here, we propose and design a scaffolding nanoparticle (NP) system that enables us to sample cytoplasmic contents without rupturing the cellular membrane, by mimicking the unusual features of clathrin. Our simulation results reveal the design principles, following which scaffolding NPs can extract lipid vesicles from plasma membranes, with both the pathway and the mechanism resembling the clathrin-mediated endocytosis, i.e. multiple NPs deposit at the membrane, assembling into cage-like structures to deform the membrane into a vesicle shape. As important design parameters, the interaction between different NPs should be properly stronger than that between each NP and the membrane to ensure the cage formation, and optimal NP concentration and the membrane surface tension are also requisite for extracting lipid vesicles. Our results provide useful guidelines for design of bio-inspired scaffolding NPs as an intelligent machine for practical use in but not limited to the single-cell analysis.
Collapse
|
37
|
Gan Q, Watanabe S. Synaptic Vesicle Endocytosis in Different Model Systems. Front Cell Neurosci 2018; 12:171. [PMID: 30002619 PMCID: PMC6031744 DOI: 10.3389/fncel.2018.00171] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 06/01/2018] [Indexed: 11/13/2022] Open
Abstract
Neurotransmission in complex animals depends on a choir of functionally distinct synapses releasing neurotransmitters in a highly coordinated manner. During synaptic signaling, vesicles fuse with the plasma membrane to release their contents. The rate of vesicle fusion is high and can exceed the rate at which synaptic vesicles can be re-supplied by distant sources. Thus, local compensatory endocytosis is needed to replenish the synaptic vesicle pools. Over the last four decades, various experimental methods and model systems have been used to study the cellular and molecular mechanisms underlying synaptic vesicle cycle. Clathrin-mediated endocytosis is thought to be the predominant mechanism for synaptic vesicle recycling. However, recent studies suggest significant contribution from other modes of endocytosis, including fast compensatory endocytosis, activity-dependent bulk endocytosis, ultrafast endocytosis, as well as kiss-and-run. Currently, it is not clear whether a universal model of vesicle recycling exist for all types of synapses. It is possible that each synapse type employs a particular mode of endocytosis. Alternatively, multiple modes of endocytosis operate at the same synapse, and the synapse toggles between different modes depending on its activity level. Here we compile review and research articles based on well-characterized model systems: frog neuromuscular junctions, C. elegans neuromuscular junctions, Drosophila neuromuscular junctions, lamprey reticulospinal giant axons, goldfish retinal ribbon synapses, the calyx of Held, and rodent hippocampal synapses. We will compare these systems in terms of their known modes and kinetics of synaptic vesicle endocytosis, as well as the underlying molecular machineries. We will also provide the future development of this field.
Collapse
Affiliation(s)
- Quan Gan
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shigeki Watanabe
- Department of Cell Biology, 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
| |
Collapse
|
38
|
Milosevic I. Revisiting the Role of Clathrin-Mediated Endoytosis in Synaptic Vesicle Recycling. Front Cell Neurosci 2018; 12:27. [PMID: 29467622 PMCID: PMC5807904 DOI: 10.3389/fncel.2018.00027] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/17/2018] [Indexed: 12/11/2022] Open
Abstract
Without robust mechanisms to efficiently form new synaptic vesicles (SVs), the tens to hundreds of SVs typically present at the neuronal synapse would be rapidly used up, even at modest levels of neuronal activity. SV recycling is thus critical for synaptic physiology and proper function of sensory and nervous systems. Yet, more than four decades after it was originally proposed that the SVs are formed and recycled locally at the presynaptic terminals, the mechanisms of endocytic processes at the synapse are heavily debated. Clathrin-mediated endocytosis, a type of endocytosis that capitalizes on the clathrin coat, a number of adaptor and accessory proteins, and the GTPase dynamin, is well understood, while the contributions of clathrin-independent fast endocytosis, kiss-and-run, bulk endocytosis and ultrafast endocytosis are still being evaluated. This review article revisits and summarizes the current knowledge on the SV reformation with a focus on clathrin-mediated endocytosis, and it discusses the modes of SV formation from endosome-like structures at the synapse. Given the importance of this topic, future advances in this active field are expected to contribute to better comprehension of neurotransmission, and to have general implications for neuroscience and medicine.
Collapse
Affiliation(s)
- Ira Milosevic
- Synaptic Vesicle Dynamics Group, European Neuroscience Institute (ENI), University Medical Center Göttingen (UMG), Göttingen, Germany
| |
Collapse
|
39
|
Brereton E, Fassi E, Araujo GC, Dodd J, Telegrafi A, Pathak SJ, Shinawi M. Mutations in the PH Domain of DNM1 are associated with a nonepileptic phenotype characterized by developmental delay and neurobehavioral abnormalities. Mol Genet Genomic Med 2018; 6:294-300. [PMID: 29397573 PMCID: PMC5902389 DOI: 10.1002/mgg3.362] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 11/13/2017] [Accepted: 11/15/2017] [Indexed: 11/30/2022] Open
Abstract
Background Dynamin 1 is a protein involved in the synaptic vesicle cycle, which facilitates the exocytosis of neurotransmitters necessary for normal signaling and development in the central nervous system. Pathogenic variants in DNM1 have been implicated in global developmental delay (DD), severe intellectual disability (ID), and notably, epileptic encephalopathy. All previously reported DNM1 pathogenic variants causing this severe phenotype occur in the GTPase and Middle domains of the dynamin 1 protein. Methods We used whole‐exome sequencing to characterize the molecular basis of DD and autistic symptoms in two identical siblings. Results The twin siblings exhibit mild to moderate ID and autistic symptoms but no epileptic encephalopathy. Exome sequencing revealed a genetic variant, c.1603A>G (p.Lys535Glu), in the PH domain of dynamin 1. Previous in vitro studies showed that mutations at Lys535 inhibit endocytosis and impair PH loop binding to PIP2. Conclusions Our data suggest a previously undescribed milder phenotype associated with a missense genetic variant in the PH domain of dynamin 1.
Collapse
Affiliation(s)
- Emily Brereton
- Washington University School of Medicine, St. Louis, MO, USA
| | - Emily Fassi
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Gabriel C Araujo
- Department of Psychology, St Louis Children's Hospital, St. Louis, MO, USA
| | - Jonathan Dodd
- Department of Psychology, St Louis Children's Hospital, St. Louis, MO, USA
| | | | - Sheel J Pathak
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Marwan Shinawi
- Washington University School of Medicine, St. Louis, MO, USA.,Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
40
|
Li H, Santos MS, Park CK, Dobry Y, Voglmaier SM. VGLUT2 Trafficking Is Differentially Regulated by Adaptor Proteins AP-1 and AP-3. Front Cell Neurosci 2017; 11:324. [PMID: 29123471 PMCID: PMC5662623 DOI: 10.3389/fncel.2017.00324] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 09/28/2017] [Indexed: 01/25/2023] Open
Abstract
Release of the major excitatory neurotransmitter glutamate by synaptic vesicle exocytosis depends on glutamate loading into synaptic vesicles by vesicular glutamate transporters (VGLUTs). The two principal isoforms, VGLUT1 and 2, exhibit a complementary pattern of expression in adult brain that broadly distinguishes cortical (VGLUT1) and subcortical (VGLUT2) systems, and correlates with distinct physiological properties in synapses expressing these isoforms. Differential trafficking of VGLUT1 and 2 has been suggested to underlie their functional diversity. Increasing evidence suggests individual synaptic vesicle proteins use specific sorting signals to engage specialized biochemical mechanisms to regulate their recycling. We observed that VGLUT2 recycles differently in response to high frequency stimulation than VGLUT1. Here we further explore the trafficking of VGLUT2 using a pHluorin-based reporter, VGLUT2-pH. VGLUT2-pH exhibits slower rates of both exocytosis and endocytosis than VGLUT1-pH. VGLUT2-pH recycling is slower than VGLUT1-pH in both hippocampal neurons, which endogenously express mostly VGLUT1, and thalamic neurons, which endogenously express mostly VGLUT2, indicating that protein identity, not synaptic vesicle membrane or neuronal cell type, controls sorting. We characterize sorting signals in the C-terminal dileucine-like motif, which plays a crucial role in VGLUT2 trafficking. Disruption of this motif abolishes synaptic targeting of VGLUT2 and essentially eliminates endocytosis of the transporter. Mutational and biochemical analysis demonstrates that clathrin adaptor proteins (APs) interact with VGLUT2 at the dileucine-like motif. VGLUT2 interacts with AP-2, a well-studied adaptor protein for clathrin mediated endocytosis. In addition, VGLUT2 also interacts with the alternate adaptors, AP-1 and AP-3. VGLUT2 relies on distinct recycling mechanisms from VGLUT1. Abrogation of these differences by pharmacological and molecular inhibition reveals that these mechanisms are dependent on the adaptor proteins AP-1 and AP-3. Further, shRNA-mediated knockdown reveals differential roles for AP-1 and AP-3 in VGLUT2 recycling.
Collapse
Affiliation(s)
- Haiyan Li
- Department of Psychiatry, School of Medicine, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, United States
| | - Magda S Santos
- Department of Psychiatry, School of Medicine, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, United States
| | - Chihyung K Park
- Department of Psychiatry, School of Medicine, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, United States
| | - Yuriy Dobry
- Department of Psychiatry, School of Medicine, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, United States
| | - Susan M Voglmaier
- Department of Psychiatry, School of Medicine, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
41
|
Kong Y, Hanna MES, Zhuo D, Chang KG, Bozorg-Grayeli T, Melosh NA. Self-Assembly of Mesoscale Artificial Clathrin Mimics. ACS NANO 2017; 11:9889-9897. [PMID: 28921943 DOI: 10.1021/acsnano.7b03739] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Fluidic control and sampling in complex environments is an important process in biotechnology, materials synthesis, and microfluidics. An elegant solution to this problem has evolved in nature through cellular endocytosis, where the dynamic recruitment, self-assembly, and spherical budding of clathrin proteins allows cells to sample their external environment. Yet despite the importance and utility of endocytosis, artificial systems which can replicate this dynamic behavior have not been developed. Guided by clathrin's unusual structure, we created simplified metallic microparticles that capture the three-legged shape, particle curvature, and interfacial attachment characteristics of clathrin. These artificial clathrin mimics successfully recreate biomimetic analogues of clathrin's recruitment, assembly, and budding, ultimately forming extended networks at fluid interfaces and invaginating immiscible phases into spheres under external fields. Particle curvature was discovered to be a critical structural motif, greatly limiting irreversible aggregation and inducing the legs' selective tip-to-tip attraction. This architecture provides a template for a class of active self-assembly units to drive structural and dimensional transformations of liquid-liquid interfaces and microscale fluidic sampling.
Collapse
Affiliation(s)
- Yifan Kong
- Department of Materials Science and Engineering, Stanford University , 476 Lomita Mall, Stanford, California 94305, United States
| | - Mina-Elraheb S Hanna
- Department of Materials Science and Engineering, Stanford University , 476 Lomita Mall, Stanford, California 94305, United States
| | - Denys Zhuo
- Department of Materials Science and Engineering, Stanford University , 476 Lomita Mall, Stanford, California 94305, United States
| | - Katherine G Chang
- Department of Materials Science and Engineering, Stanford University , 476 Lomita Mall, Stanford, California 94305, United States
| | - Tara Bozorg-Grayeli
- Department of Materials Science and Engineering, Stanford University , 476 Lomita Mall, Stanford, California 94305, United States
| | - Nicholas A Melosh
- Department of Materials Science and Engineering, Stanford University , 476 Lomita Mall, Stanford, California 94305, United States
| |
Collapse
|
42
|
Rampérez A, Sánchez-Prieto J, Torres M. Brefeldin A sensitive mechanisms contribute to endocytotic membrane retrieval and vesicle recycling in cerebellar granule cells. J Neurochem 2017; 141:662-675. [PMID: 28295320 DOI: 10.1111/jnc.14017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 02/22/2017] [Accepted: 03/01/2017] [Indexed: 11/26/2022]
Abstract
The recycling of synaptic vesicle (SV) proteins and transmitter release occur at multiple sites along the axon. These processes are sensitive to inhibition of the small GTP binding protein ARF1, which regulates the adaptor protein 1 and 3 complex (AP-1/AP-3). As the axon matures, SV recycling becomes restricted to the presynaptic bouton, and its machinery undergoes a complex process of maturation. We used the styryl dye FM1-43 to highlight differences in the efficiency of membrane recycling at different sites in cerebellar granule cells cultured for 7 days in vitro. We used Brefeldin A (BFA) to inhibit AP-1/AP-3-mediated recycling and to test the contribution of this pathway to the heterogeneity of the responses when these cells are strongly stimulated. Combining imaging techniques and ultrastructural analyses, we found a significant decrease in the density of functional boutons and an increase in the presence of endosome-like structures within the boutons of cells incubated with BFA prior to FM1-43 loading. Such effects were not observed when BFA was added 5 min after the end of the loading step, when endocytosis was almost fully completed. In this situation, vesicles were found closer to the active zone (AZ) in boutons exposed to BFA. Together, these data suggest that the AP-1/AP-3 pathway contributes to SV recycling, affecting different steps in all boutons but not equally, and thus being partly responsible for the heterogeneity of the different recycling efficiencies. Cover Image for this issue: doi. 10.1111/jnc.13801.
Collapse
Affiliation(s)
- Alberto Rampérez
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - José Sánchez-Prieto
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Magdalena Torres
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| |
Collapse
|
43
|
Li YC, Kavalali ET. Synaptic Vesicle-Recycling Machinery Components as Potential Therapeutic Targets. Pharmacol Rev 2017; 69:141-160. [PMID: 28265000 PMCID: PMC5394918 DOI: 10.1124/pr.116.013342] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Presynaptic nerve terminals are highly specialized vesicle-trafficking machines. Neurotransmitter release from these terminals is sustained by constant local recycling of synaptic vesicles independent from the neuronal cell body. This independence places significant constraints on maintenance of synaptic protein complexes and scaffolds. Key events during the synaptic vesicle cycle-such as exocytosis and endocytosis-require formation and disassembly of protein complexes. This extremely dynamic environment poses unique challenges for proteostasis at synaptic terminals. Therefore, it is not surprising that subtle alterations in synaptic vesicle cycle-associated proteins directly or indirectly contribute to pathophysiology seen in several neurologic and psychiatric diseases. In contrast to the increasing number of examples in which presynaptic dysfunction causes neurologic symptoms or cognitive deficits associated with multiple brain disorders, synaptic vesicle-recycling machinery remains an underexplored drug target. In addition, irrespective of the involvement of presynaptic function in the disease process, presynaptic machinery may also prove to be a viable therapeutic target because subtle alterations in the neurotransmitter release may counter disease mechanisms, correct, or compensate for synaptic communication deficits without the need to interfere with postsynaptic receptor signaling. In this article, we will overview critical properties of presynaptic release machinery to help elucidate novel presynaptic avenues for the development of therapeutic strategies against neurologic and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Ying C Li
- Departments of Neuroscience (Y.C.L., E.T.K.) and Physiology (E.T.K.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ege T Kavalali
- Departments of Neuroscience (Y.C.L., E.T.K.) and Physiology (E.T.K.), University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
44
|
RA Differentiation Enhances Dopaminergic Features, Changes Redox Parameters, and Increases Dopamine Transporter Dependency in 6-Hydroxydopamine-Induced Neurotoxicity in SH-SY5Y Cells. Neurotox Res 2017; 31:545-559. [DOI: 10.1007/s12640-016-9699-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 12/28/2016] [Accepted: 12/30/2016] [Indexed: 12/19/2022]
|
45
|
Synaptic Vesicle Endocytosis Occurs on Multiple Timescales and Is Mediated by Formin-Dependent Actin Assembly. Neuron 2017; 93:854-866.e4. [DOI: 10.1016/j.neuron.2017.02.011] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 12/12/2016] [Accepted: 01/23/2017] [Indexed: 11/21/2022]
|
46
|
Tham DKL, Joshi B, Moukhles H. Aquaporin-4 Cell-Surface Expression and Turnover Are Regulated by Dystroglycan, Dynamin, and the Extracellular Matrix in Astrocytes. PLoS One 2016; 11:e0165439. [PMID: 27788222 PMCID: PMC5082936 DOI: 10.1371/journal.pone.0165439] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 10/11/2016] [Indexed: 11/19/2022] Open
Abstract
The water-permeable channel aquaporin-4 (AQP4) is highly expressed in perivascular astrocytes of the mammalian brain and represents the major conduit for water across the blood-brain barrier. Within these cells, AQP4 is found in great quantities at perivascular endfoot sites but is detected in lesser amounts at the membrane domains within the brain parenchyma. We had previously established that this polarization was regulated by the interaction between dystroglycan (DG), an extracellular matrix receptor that is co-expressed with AQP4, and the laminin that is contained within the perivascular basal lamina. In the present study, we have attempted to describe the mechanisms that underlie this regulation, using primary astrocyte cultures. Via biotinylation, we found that the cell-surface expression of AQP4 is DG-dependent and is potentiated by laminin. We also determined that this laminin-dependent increase occurs not through an upregulation of total AQP4 levels, but rather from a redirection of AQP4 from an intracellular, EEA-1-associated pool to the cell surface. We then demonstrated an association between DG and dynamin and showed that dynamin functioned in conjunction with clathrin to regulate surface AQP4 amounts. Furthermore, we observed that DG preferentially binds to the inactive forms of dynamin, suggesting that this interaction was inhibitory for AQP4 endocytosis. Finally, we showed that laminin selectively upregulates the cell-surface expression of the M23 isoform of AQP4. Our data therefore indicate that the dual interation of DG with laminin and dynamin is involved in the regulation of AQP4 internalization, leading to its asymmetric enrichment at perivascular astrocyte endfeet.
Collapse
Affiliation(s)
- Daniel Kai Long Tham
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Bharat Joshi
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hakima Moukhles
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
47
|
Meunier FA, Gutiérrez LM. Captivating New Roles of F-Actin Cortex in Exocytosis and Bulk Endocytosis in Neurosecretory Cells. Trends Neurosci 2016; 39:605-613. [DOI: 10.1016/j.tins.2016.07.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/01/2016] [Accepted: 07/06/2016] [Indexed: 12/01/2022]
|
48
|
Modes and mechanisms of synaptic vesicle recycling. Curr Opin Neurobiol 2016; 39:17-23. [DOI: 10.1016/j.conb.2016.03.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/02/2016] [Accepted: 03/07/2016] [Indexed: 11/30/2022]
|
49
|
Werner C, Pauli M, Doose S, Weishaupt A, Haselmann H, Grünewald B, Sauer M, Heckmann M, Toyka KV, Asan E, Sommer C, Geis C. Human autoantibodies to amphiphysin induce defective presynaptic vesicle dynamics and composition. Brain 2015; 139:365-79. [DOI: 10.1093/brain/awv324] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 09/25/2015] [Indexed: 12/16/2022] Open
Abstract
Abstract
See Irani (doi:10.1093/awv364) for a scientific commentary on this article.
Stiff-person syndrome is the prototype of a central nervous system disorder with autoantibodies targeting presynaptic antigens. Patients with paraneoplastic stiff-person syndrome may harbour autoantibodies to the BAR (Bin/Amphiphysin/Rvs) domain protein amphiphysin, which target its SH3 domain. These patients have neurophysiological signs of compromised central inhibition and respond to symptomatic treatment with medication enhancing GABAergic transmission. High frequency neurotransmission as observed in tonic GABAergic interneurons relies on fast exocytosis of neurotransmitters based on compensatory endocytosis. As amphiphysin is involved in clathrin-mediated endocytosis, patient autoantibodies are supposed to interfere with this function, leading to disinhibition by reduction of GABAergic neurotransmission. We here investigated the effects of human anti-amphiphysin autoantibodies on structural components of presynaptic boutons ex vivo and in vitro using electron microscopy and super-resolution direct stochastic optical reconstruction microscopy. Ultrastructural analysis of spinal cord presynaptic boutons was performed after in vivo intrathecal passive transfer of affinity-purified human anti-amphiphysin autoantibodies in rats and revealed signs of markedly disabled clathrin-mediated endocytosis. This was unmasked at high synaptic activity and characterized by a reduction of the presynaptic vesicle pool, clathrin coated intermediates, and endosome-like structures. Super-resolution microscopy of inhibitory GABAergic presynaptic boutons in primary neurons revealed that specific human anti-amphiphysin immunoglobulin G induced an increase of the essential vesicular protein synaptobrevin 2 and a reduction of synaptobrevin 7. This constellation suggests depletion of resting pool vesicles and trapping of releasable pool vesicular proteins at the plasma membrane. Similar effects were found in amphiphysin-deficient neurons from knockout mice. Application of specific patient antibodies did not show additional effects. Blocking alternative pathways of clathrin-independent endocytosis with brefeldin A reversed the autoantibody induced effects on molecular vesicle composition. Endophilin as an interaction partner of amphiphysin showed reduced clustering within presynaptic terminals. Collectively, these results point towards an autoantibody-induced structural disorganization in GABAergic synapses with profound changes in presynaptic vesicle pools, activation of alternative endocytic pathways, and potentially compensatory rearrangement of proteins involved in clathrin-mediated endocytosis. Our findings provide novel insights into synaptic pathomechanisms in a prototypic antibody-mediated central nervous system disease, which may serve as a proof-of-principle example in this evolving group of autoimmune disorders associated with autoantibodies to synaptic antigens.
Collapse
Affiliation(s)
- Christian Werner
- 1 Hans-Berger Department of Neurology, Jena University Hospital, Erlanger Allee 101, 07747 Jena, Germany
- 2 Department of Neurology, University of Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany
| | - Martin Pauli
- 3 Department of Neurophysiology, Institute of Physiology, University of Würzburg, Roentgenring 9, 97070 Würzburg, Germany
| | - Sören Doose
- 4 Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Andreas Weishaupt
- 2 Department of Neurology, University of Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany
| | - Holger Haselmann
- 1 Hans-Berger Department of Neurology, Jena University Hospital, Erlanger Allee 101, 07747 Jena, Germany
- 2 Department of Neurology, University of Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany
- 5 Center for Sepsis Control and Care (CSCC), Jena University Hospital, Erlanger Allee 101, 07747 Jena, Germany
| | - Benedikt Grünewald
- 1 Hans-Berger Department of Neurology, Jena University Hospital, Erlanger Allee 101, 07747 Jena, Germany
- 2 Department of Neurology, University of Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany
- 5 Center for Sepsis Control and Care (CSCC), Jena University Hospital, Erlanger Allee 101, 07747 Jena, Germany
| | - Markus Sauer
- 4 Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Manfred Heckmann
- 3 Department of Neurophysiology, Institute of Physiology, University of Würzburg, Roentgenring 9, 97070 Würzburg, Germany
| | - Klaus V. Toyka
- 2 Department of Neurology, University of Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany
| | - Esther Asan
- 6 Institute for Anatomy and Cell Biology, University of Würzburg, Koellikerstrasse 6, 97070 Würzburg, Germany
| | - Claudia Sommer
- 2 Department of Neurology, University of Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany
| | - Christian Geis
- 1 Hans-Berger Department of Neurology, Jena University Hospital, Erlanger Allee 101, 07747 Jena, Germany
- 2 Department of Neurology, University of Würzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany
- 5 Center for Sepsis Control and Care (CSCC), Jena University Hospital, Erlanger Allee 101, 07747 Jena, Germany
| |
Collapse
|
50
|
Abstract
In the CNS (central nervous system), nerve cells communicate by transmitting signals from one to the next across chemical synapses. Electrical signals trigger controlled secretion of neurotransmitter by exocytosis of SV (synaptic vesicles) at the presynaptic site. Neurotransmitters diffuse across the synaptic cleft, activate receptor channels in the receiving neuron at the postsynaptic site, and thereby elicit a new electrical signal. Repetitive stimulation should result in fast depletion of fusion-competent SVs, given their limited number in the presynaptic bouton. Therefore, to support repeated rounds of release, a fast trafficking cycle is required that couples exocytosis and compensatory endocytosis. During this exo-endocytic cycle, a defined stoichiometry of SV proteins has to be preserved, that is, membrane proteins have to be sorted precisely. However, how this sorting is accomplished on a molecular level is poorly understood. In the present chapter we review recent findings regarding the molecular composition of SVs and the mechanisms that sort SV proteins during compensatory endocytosis. We identify self-assembly of SV components and individual cargo recognition by sorting adaptors as major mechanisms for maintenance of the SV protein complement.
Collapse
|