1
|
Chhabra A, Hoffmann C, Pérez GA, Korobeinikov AA, Rentsch J, Hümpfer N, Kokwaro L, Gnidovec L, Petrovic A, Wallace JN, Tromm JV, Román-Vendrell C, Johnson EC, Ranković B, Perego E, Volpi T, Fernández-Busnadiego R, Köster S, Rizzoli SO, Ewers H, Morgan JR, Milovanovic D. Condensates of synaptic vesicles and synapsin are molecular beacons for actin sequestering and polymerization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.19.604346. [PMID: 39071264 PMCID: PMC11275919 DOI: 10.1101/2024.07.19.604346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Neuronal communication relies on precisely maintained synaptic vesicle (SV) clusters, which assemble via liquid-liquid phase separation (LLPS). This process requires synapsins, the major synaptic phosphoproteins, which are known to bind actin. The reorganization of SVs, synapsins and actin is a hallmark of synaptic activity, but their interplay is still unclear. Here, we combined the reconstitution approaches, expansion microscopy, super-resolution imaging and cryo-electron tomography to dissect the roles of synapsin-SV condensates in the organization of the presynaptic actin cytoskeleton. Our data indicate that LLPS of synapsin initiates actin polymerization, allowing for SV:synapsin:actin assemblies to facilitate the mesoscale organization of SV clusters along axons mimicking the native presynaptic organization in both lamprey and mammalian synapses. Understanding the relationship between the actin network and synapsin-SVs condensates is an essential building block on a roadmap to unravel how coordinated neurotransmission along the axon enables circuit function and behavior.
Collapse
Affiliation(s)
- Akshita Chhabra
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Whitman Center, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
| | - Christian Hoffmann
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Whitman Center, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
| | - Gerard Aguilar Pérez
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Whitman Center, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
| | - Aleksandr A. Korobeinikov
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | - Jakob Rentsch
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Nadja Hümpfer
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Linda Kokwaro
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Einstein Center for Neuroscience, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Luka Gnidovec
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | - Arsen Petrovic
- Institute for Neuropathology, University Medical Center Göttingen, 37073, Göttingen, Germany
| | - Jaqulin N. Wallace
- The Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
| | - Johannes Vincent Tromm
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Whitman Center, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
- The Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
| | - Cristina Román-Vendrell
- The Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
| | - Emma C. Johnson
- The Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
| | - Branislava Ranković
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Whitman Center, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
| | - Eleonora Perego
- Institute for X-Ray Physics, University of Göttingen, Friedrich-Hund-Platz 1, 37077 Göttingen, Germany
| | - Tommaso Volpi
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | - Sarah Köster
- Institute for X-Ray Physics, University of Göttingen, Friedrich-Hund-Platz 1, 37077 Göttingen, Germany
| | - Silvio O. Rizzoli
- Institute for Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073, Göttingen, Germany
| | - Helge Ewers
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Jennifer R. Morgan
- The Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
| | - Dragomir Milovanovic
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Whitman Center, Marine Biological Laboratory, 02543 Woods Hole, MA, USA
- Einstein Center for Neuroscience, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| |
Collapse
|
2
|
Earnhardt-San AL, Baker EC, Cilkiz KZ, Cardoso RC, Ghaffari N, Long CR, Riggs PK, Randel RD, Riley DG, Welsh TH. Evaluation of Prenatal Transportation Stress on DNA Methylation (DNAm) and Gene Expression in the Hypothalamic-Pituitary-Adrenal (HPA) Axis Tissues of Mature Brahman Cows. Genes (Basel) 2025; 16:191. [PMID: 40004522 PMCID: PMC11855312 DOI: 10.3390/genes16020191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/27/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: The experience of prenatal stress results in various physiological disorders due to an alteration of an offspring's methylome and transcriptome. The objective of this study was to determine whether PNS affects DNA methylation (DNAm) and gene expression in the stress axis tissues of mature Brahman cows. Methods: Samples were collected from the paraventricular nucleus (PVN), anterior pituitary (PIT), and adrenal cortex (AC) of 5-year-old Brahman cows that were prenatally exposed to either transportation stress (PNS, n = 6) or were not transported (Control, n = 8). The isolated DNA and RNA samples were, respectively, used for methylation and RNA-Seq analyses. A gene ontology and KEGG pathway enrichment analysis of each data set within each sample tissue was conducted with the DAVID Functional Annotation Tool. Results: The DNAm analysis revealed 3, 64, and 99 hypomethylated and 2, 93, and 90 hypermethylated CpG sites (FDR < 0.15) within the PVN, PIT, and AC, respectively. The RNA-Seq analysis revealed 6, 25, and 5 differentially expressed genes (FDR < 0.15) in the PVN, PIT, and AC, respectively, that were up-regulated in the PNS group relative to the Control group, as well as 24 genes in the PIT that were down-regulated. Based on the enrichment analysis, several developmental and cellular processes, such as maintenance of the actin cytoskeleton, cell motility, signal transduction, neurodevelopment, and synaptic function, were potentially modulated. Conclusions: The methylome and transcriptome were altered in the stress axis tissues of mature cows that had been exposed to prenatal transportation stress. These findings are relevant to understanding how prenatal experiences may affect postnatal neurological functions.
Collapse
Affiliation(s)
- Audrey L. Earnhardt-San
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
- Texas A&M AgriLife Research Center, Overton, TX 75684, USA
| | - Emilie C. Baker
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Kubra Z. Cilkiz
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Rodolfo C. Cardoso
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Noushin Ghaffari
- Department of Computer Science, Prairie View A&M University, Prairie View, TX 77070, USA;
| | - Charles R. Long
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
- Department of Computer Science, Prairie View A&M University, Prairie View, TX 77070, USA;
| | - Penny K. Riggs
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Ronald D. Randel
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
- Department of Computer Science, Prairie View A&M University, Prairie View, TX 77070, USA;
| | - David G. Riley
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Thomas H. Welsh
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| |
Collapse
|
3
|
Mufti K, Cordova M, Scott EN, Trueman JN, Lovnicki JM, Loucks CM, Rassekh SR, Ross CJD, Carleton BC. Genomic variations associated with risk and protection against vincristine-induced peripheral neuropathy in pediatric cancer patients. NPJ Genom Med 2024; 9:56. [PMID: 39500896 PMCID: PMC11538333 DOI: 10.1038/s41525-024-00443-7] [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: 01/23/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024] Open
Abstract
Vincristine-induced peripheral neuropathy is a common and highly debilitating toxicity from vincristine treatment that affects quality of life and often requires dose reduction, potentially affecting survival. Although previous studies demonstrated genetic factors are associated with vincristine neuropathy risk, the clinical relevance of most identified variants is limited by small sample sizes and unclear clinical phenotypes. A genome-wide association study was conducted in 1100 cases and controls matched by vincristine dose and genetic ancestry, uncovering a statistically significant (p < 5.0 × 10-8) variant in MCM3AP gene that substantially increases the risk of neuropathy and 12 variants protective against neuropathy within/near SPDYA, METTL8, PDE4D, FBN2, ZFAND3, NFIB, PAPPA, LRRTM3, NRG3, VTI1A, ARHGAP5, and ACTN1. A follow-up pathway analysis reveals the involvement of four key pathways, including nerve structure and development, myelination, neuronal transmission, and cytoskeleton/microfibril function pathways. These findings present potential actionable genomic markers of vincristine neuropathy and offer opportunities for tailored interventions to improve vincristine safety in children with cancer. This study is registered with ClinicalTrials.gov under the title National Active Surveillance Network and Pharmacogenomics of Adverse Drug Reactions in Children (ID NCT00414115, registered on December 21, 2006).
Collapse
Affiliation(s)
- Kheireddin Mufti
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Miguel Cordova
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Erika N Scott
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jessica N Trueman
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Pharmaceutical Outcomes Programme, BC Children's Hospital, Vancouver, BC, Canada
| | - Jessica M Lovnicki
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
- Pharmaceutical Outcomes Programme, BC Children's Hospital, Vancouver, BC, Canada
| | - Catrina M Loucks
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Anesthesiology, Pharmacology & Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Shahrad R Rassekh
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
- Division of Hematology, Oncology & Bone Marrow Transplant, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Colin J D Ross
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada.
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada.
| | - Bruce C Carleton
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada.
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
- Pharmaceutical Outcomes Programme, BC Children's Hospital, Vancouver, BC, Canada.
| |
Collapse
|
4
|
Micovic K, Canuel A, Remtulla A, Chuyen A, Byrsan M, McGarry DJ, Olson MF. Mical1 deletion in tyrosinase expressing cells affects mouse running gaits. GENES, BRAIN, AND BEHAVIOR 2024; 23:e70004. [PMID: 39344934 PMCID: PMC11440367 DOI: 10.1111/gbb.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 10/01/2024]
Abstract
Neuronal development is a highly regulated process that is dependent on the correct coordination of cellular responses to extracellular cues. In response to semaphorin axon guidance proteins, the MICAL1 protein is stimulated to produce reactive oxygen species that oxidize actin on specific methionine residues, leading to filamentous actin depolymerization and consequent changes in neuronal growth cone dynamics. Crossing genetically modified mice homozygous for floxed Mical1 (Mical1fl/fl) alleles with transgenic mice expressing Cre recombinase under the control of a tyrosinase gene enhancer/promoter (Tyr::Cre) enabled conditional Mical1 deletion. Immunohistochemical analysis showed Mical1 expression in the cerebellum, which plays a prominent role in the coordination of motor movements, with reduced Mical1 expression in Mical1fl/fl mice co-expressing Tyr::Cre. Analysis of the gaits of mice running on a treadmill showed that both male and female Mical1fl/fl, Tyr::Cre mutant mice had significant alterations to their striding patterns relative to wild-type mice, although the specific aspects of their altered gaits differed between the sexes. Additional motor tests that involved movement on a rotating rod, descending a vertical pole, or crossing a balance beam did not show significant differences between the genotypes, suggesting that the effect of the Mical1fl/fl, Tyr::Cre genetic modifications was only manifested during specific highly coordinated movements that contribute to running. These findings indicate that there is a behavioral consequence in Mical1fl/fl, Tyr::Cre mutant mice that affects motor control as manifested by alterations in their gait.
Collapse
Affiliation(s)
- Katarina Micovic
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
| | - Alicia Canuel
- Department of Pharmacology and ToxicologyUniversity of TorontoTorontoOntarioCanada
| | - Aasiya Remtulla
- Department of Pharmacology and ToxicologyUniversity of TorontoTorontoOntarioCanada
| | - Alexandre Chuyen
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
| | - Margarita Byrsan
- Biomedical Engineering ProgramToronto Metropolitan UniversityTorontoOntarioCanada
| | - David J. McGarry
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
| | - Michael F. Olson
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
- Department of Pharmacology and ToxicologyUniversity of TorontoTorontoOntarioCanada
- Biomedical Engineering ProgramToronto Metropolitan UniversityTorontoOntarioCanada
| |
Collapse
|
5
|
Wang J, Wu M, Magupalli VG, Dahlberg PD, Wu H, Jensen GJ. Human NLRP3 inflammasome activation leads to formation of condensate at the microtubule organizing center. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612739. [PMID: 39314395 PMCID: PMC11419111 DOI: 10.1101/2024.09.12.612739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The NLRP3 inflammasome is a multi-protein molecular machine that mediates inflammatory responses in innate immunity. Its dysregulation has been linked to a large number of human diseases. Using cryogenic fluorescence-guided focused-ion-beam (cryo-FIB) milling and electron cryo-tomography (cryo-ET), we obtained 3-D images of the NLRP3 inflammasome in situ at various stages of its activation at macromolecular resolution. The cryo-tomograms unexpectedly reveal dense condensates of the human macrophage NLRP3 inflammasome that form within and around the microtubule organizing center (MTOC). We also find that following activation, the trans-Golgi network disperses and 50-nm NLRP3-associated vesicles appear which likely ferry NLRP3 to the MTOC. At later time points after activation, the electron-dense condensates progressively solidify and the cells undergo pyroptosis with widespread damaged mitochondria and autophagasomal structures.
Collapse
Affiliation(s)
- Jue Wang
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125
- Division of CryoEM and Bioimaging, SSRL, SLAC National Accelerator Laboratory, Menlo Park, CA 94025
| | - Man Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston MA 02446
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston MA 02446
| | - Venkat G Magupalli
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston MA 02446
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston MA 02446
| | - Peter D Dahlberg
- Division of CryoEM and Bioimaging, SSRL, SLAC National Accelerator Laboratory, Menlo Park, CA 94025
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston MA 02446
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston MA 02446
| | - Grant J Jensen
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602
| |
Collapse
|
6
|
Kim J, Bustamante E, Sotonyi P, Maxwell N, Parameswaran P, Kent JK, Wetsel WC, Soderblom EJ, Rácz B, Soderling SH. Presynaptic Rac1 in the hippocampus selectively regulates working memory. eLife 2024; 13:RP97289. [PMID: 39046788 PMCID: PMC11268886 DOI: 10.7554/elife.97289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024] Open
Abstract
One of the most extensively studied members of the Ras superfamily of small GTPases, Rac1 is an intracellular signal transducer that remodels actin and phosphorylation signaling networks. Previous studies have shown that Rac1-mediated signaling is associated with hippocampal-dependent working memory and longer-term forms of learning and memory and that Rac1 can modulate forms of both pre- and postsynaptic plasticity. How these different cognitive functions and forms of plasticity mediated by Rac1 are linked, however, is unclear. Here, we show that spatial working memory in mice is selectively impaired following the expression of a genetically encoded Rac1 inhibitor at presynaptic terminals, while longer-term cognitive processes are affected by Rac1 inhibition at postsynaptic sites. To investigate the regulatory mechanisms of this presynaptic process, we leveraged new advances in mass spectrometry to identify the proteomic and post-translational landscape of presynaptic Rac1 signaling. We identified serine/threonine kinases and phosphorylated cytoskeletal signaling and synaptic vesicle proteins enriched with active Rac1. The phosphorylated sites in these proteins are at positions likely to have regulatory effects on synaptic vesicles. Consistent with this, we also report changes in the distribution and morphology of synaptic vesicles and in postsynaptic ultrastructure following presynaptic Rac1 inhibition. Overall, this study reveals a previously unrecognized presynaptic role of Rac1 signaling in cognitive processes and provides insights into its potential regulatory mechanisms.
Collapse
Affiliation(s)
- Jaebin Kim
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
| | - Edwin Bustamante
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
| | - Peter Sotonyi
- Department of Anatomy and Histology, University of Veterinary MedicineBudapestHungary
| | - Nicholas Maxwell
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
| | - Pooja Parameswaran
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
| | - Julie K Kent
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
| | - William C Wetsel
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
- Department of Psychiatry and Behavioral Sciences, Duke University School of MedicineDurhamUnited States
- Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University School of MedicineDurhamUnited States
- Department of Neurobiology, Duke University School of MedicineDurhamUnited States
| | - Erik J Soderblom
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
- Proteomics and Metabolomics Shared Resource and Center for Genomic and Computational Biology, Duke University School of MedicineDurhamUnited States
| | - Bence Rácz
- Department of Anatomy and Histology, University of Veterinary MedicineBudapestHungary
| | - Scott H Soderling
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
- Department of Neurobiology, Duke University School of MedicineDurhamUnited States
| |
Collapse
|
7
|
Qiu H, Wu X, Ma X, Li S, Cai Q, Ganzella M, Ge L, Zhang H, Zhang M. Short-distance vesicle transport via phase separation. Cell 2024; 187:2175-2193.e21. [PMID: 38552623 DOI: 10.1016/j.cell.2024.03.003] [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: 01/31/2023] [Revised: 01/17/2024] [Accepted: 03/02/2024] [Indexed: 04/28/2024]
Abstract
In addition to long-distance molecular motor-mediated transport, cellular vesicles also need to be moved at short distances with defined directions to meet functional needs in subcellular compartments but with unknown mechanisms. Such short-distance vesicle transport does not involve molecular motors. Here, we demonstrate, using synaptic vesicle (SV) transport as a paradigm, that phase separation of synaptic proteins with vesicles can facilitate regulated, directional vesicle transport between different presynaptic bouton sub-compartments. Specifically, a large coiled-coil scaffold protein Piccolo, in response to Ca2+ and via its C2A domain-mediated Ca2+ sensing, can extract SVs from the synapsin-clustered reserve pool condensate and deposit the extracted SVs onto the surface of the active zone protein condensate. We further show that the Trk-fused gene, TFG, also participates in COPII vesicle trafficking from ER to the ER-Golgi intermediate compartment via phase separation. Thus, phase separation may play a general role in short-distance, directional vesicle transport in cells.
Collapse
Affiliation(s)
- Hua Qiu
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Xiandeng Wu
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Xiaoli Ma
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shulin Li
- State Key Laboratory of Membrane Biology, Beijing, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China; School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qixu Cai
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Marcelo Ganzella
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, Göttingen 37077, Germany
| | - Liang Ge
- State Key Laboratory of Membrane Biology, Beijing, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China; School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Hong Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingjie Zhang
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen 518036, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
8
|
Kim J, Bustamante E, Sotonyi P, Maxwell ND, Parameswaran P, Kent JK, Wetsel WC, Soderblom EJ, Rácz B, Soderling SH. Presynaptic Rac1 in the hippocampus selectively regulates working memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.18.585488. [PMID: 38562715 PMCID: PMC10983896 DOI: 10.1101/2024.03.18.585488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
One of the most extensively studied members of the Ras superfamily of small GTPases, Rac1 is an intracellular signal transducer that remodels actin and phosphorylation signaling networks. Previous studies have shown that Rac1-mediated signaling is associated with hippocampal-dependent working memory and longer-term forms of learning and memory and that Rac1 can modulate forms of both pre- and postsynaptic plasticity. How these different cognitive functions and forms of plasticity mediated by Rac1 are linked, however, is unclear. Here, we show that spatial working memory is selectively impaired following the expression of a genetically encoded Rac1-inhibitor at presynaptic terminals, while longer-term cognitive processes are affected by Rac1 inhibition at postsynaptic sites. To investigate the regulatory mechanisms of this presynaptic process, we leveraged new advances in mass spectrometry to identify the proteomic and post-translational landscape of presynaptic Rac1 signaling. We identified serine/threonine kinases and phosphorylated cytoskeletal signaling and synaptic vesicle proteins enriched with active Rac1. The phosphorylated sites in these proteins are at positions likely to have regulatory effects on synaptic vesicles. Consistent with this, we also report changes in the distribution and morphology of synaptic vesicles and in postsynaptic ultrastructure following presynaptic Rac1 inhibition. Overall, this study reveals a previously unrecognized presynaptic role of Rac1 signaling in cognitive processes and provides insights into its potential regulatory mechanisms.
Collapse
Affiliation(s)
- Jaebin Kim
- Department of Cell Biology, Duke University Medical School, Durham, North Carolina, USA
| | - Edwin Bustamante
- Department of Cell Biology, Duke University Medical School, Durham, North Carolina, USA
| | - Peter Sotonyi
- Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, Hungary
| | - Nicholas D Maxwell
- Department of Cell Biology, Duke University Medical School, Durham, North Carolina, USA
| | - Pooja Parameswaran
- Department of Cell Biology, Duke University Medical School, Durham, North Carolina, USA
| | - Julie K Kent
- Department of Cell Biology, Duke University Medical School, Durham, North Carolina, USA
| | - William C Wetsel
- Department of Cell Biology, Duke University Medical School, Durham, North Carolina, USA
- Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University Medical School, Durham, North Carolina, USA
- Department of Neurobiology, Duke University Medical School, Durham, North Carolina, USA
| | - Erik J Soderblom
- Department of Cell Biology, Duke University Medical School, Durham, North Carolina, USA
- Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University Medical School, Durham, North Carolina, USA
| | - Bence Rácz
- Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, Hungary
| | - Scott H Soderling
- Department of Cell Biology, Duke University Medical School, Durham, North Carolina, USA
- Department of Neurobiology, Duke University Medical School, Durham, North Carolina, USA
| |
Collapse
|
9
|
Qin R, Huang L, Xu W, Qin Q, Liang X, Lai X, Huang X, Xie M, Chen L. Identification of disulfidptosis-related genes and analysis of immune infiltration characteristics in ischemic strokes. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:18939-18959. [PMID: 38052584 DOI: 10.3934/mbe.2023838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Immune infiltration plays a pivotal role in the pathogenesis of ischemic stroke. A novel form of cell death known as disulfidptosis has emerged in recent studies. However, there is currently a lack of research investigating the regulatory mechanism of disulfidptosis-related genes in immune infiltration during ischemic stroke. Using machine learning methods, we identified candidate key disulfidptosis-related genes (DRGs). Subsequently, we performed an analysis of immune cell infiltration to investigate the dysregulation of immune cells in the context of ischemic stroke. We assessed their diagnostic value by employing receiver operating characteristic (ROC) curves. To gain further insights, we conducted functional enrichment analyses to elucidate the signaling pathways associated with these seven DRGs. We identified two distinct subclusters based on the expression patterns of these seven DRGs. The unique roles of these subclusters were further evaluated through KEGG analysis and immune infiltration studies. Furthermore, we validated the expression profiles of these seven DRGs using both single-cell datasets and external datasets. Lastly, molecular docking was performed to explore potential drugs for the treatment of ischemic stroke. We identified seven DRGs. The seven DRGs are related to immune cells. Additionally, these seven DRGs also demonstrate potential diagnostic value in ischemic stroke. Functional enrichment analysis highlighted pathways such as platelet aggregation and platelet activation. Two subclusters related to disulfidptosis were defined, and functional enrichment analysis of their differentially expressed genes (DEGs) primarily involved pathways like cytokine-cytokine receptor interaction. Single-cell analysis indicated that these seven DRGs were primarily distributed among immune cell types. Molecular docking results suggested that genistein might be a potential therapeutic drug. This study has opened up new avenues for exploring the causes of ischemic stroke and developing potential therapeutic targets.
Collapse
Affiliation(s)
- Rongxing Qin
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Lijuan Huang
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
| | - Wei Xu
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
| | - Qingchun Qin
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
| | - Xiaojun Liang
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Xinyu Lai
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Xiaoying Huang
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Minshan Xie
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Li Chen
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
10
|
Fogarty MJ, Zhan WZ, Simmon VF, Vanderklish PW, Sarraf ST, Sieck GC. Novel regenerative drug, SPG302 promotes functional recovery of diaphragm muscle activity after cervical spinal cord injury. J Physiol 2023; 601:2513-2532. [PMID: 36815402 PMCID: PMC10404468 DOI: 10.1113/jp284004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
Spinal cord hemisection at C2 (C2 SH), sparing the dorsal column is widely used to investigate the effects of reduced phrenic motor neuron (PhMN) activation on diaphragm muscle (DIAm) function, with reduced DIAm activity on the injured side during eupnoea. Following C2 SH, recovery of DIAm EMG activity may occur spontaneously over subsequent days/weeks. Various strategies have been effective at improving the incidence and magnitude of DIAm recovery during eupnoea, but little is known about the effects of C2 SH on transdiaphragmatic pressure (Pdi ) during other ventilatory and non-ventilatory behaviours. We employ SPG302, a novel type of pegylated benzothiazole derivative, to assess whether enhancing synaptogenesis (i.e., enhancing spared local connections) will improve the incidence and the magnitude of recovery of DIAm EMG activity and Pdi function 14 days post-C2 SH. In anaesthetised Sprague-Dawley rats, DIAm EMG and Pdi were assessed during eupnoea, hypoxia/hypercapnia and airway occlusion prior to surgery (C2 SH or sham), immediately post-surgery and at 14 days post-surgery. In C2 SH rats, 14 days of DMSO (vehicle) or SPG302 treatments (i.p. injection) occurred. At the terminal experiment, maximum Pdi was evoked by bilateral phrenic nerve stimulation. We show that significant EMG and Pdi deficits are apparent in C2 SH compared with sham rats immediately after surgery. In C2 SH rats treated with SPG302, recovery of eupneic, hypoxia/hypercapnia and occlusion DIAm EMG was enhanced compared with vehicle rats after 14 days. Treatment with SPG302 also ameliorated Pdi deficits following C2 SH. In summary, SPG302 is an exciting new therapy to explore for use in spinal cord injuries. KEY POINTS: Despite advances in our understanding of the effects of cervical hemisection (C2 SH) on diaphragm muscle (DIAm) EMG activity, very little is understood about the impact of C2 SH on the gamut of ventilatory and non-ventilatory transdiaphragmatic pressures (Pdi ). Recovery of DIAm activity following C2 SH is improved using a variety of approaches, but very few pharmaceuticals have been shown to be effective. One way of improving DIAm recovery is to enhance the amount of latent local spared connections onto phrenic motor neurons. A novel pegylated benzothiazole derivative enhances synaptogenesis in a variety of neurodegenerative conditions. Here, using a novel therapeutic SPG302, we show that 14 days of treatment with SPG302 ameliorated DIAm EMG and Pdi deficits compared with vehicle controls. Our results show that SPG302 is a compound with very promising potential for use in improving functional outcomes post-spinal cord injury.
Collapse
Affiliation(s)
- Matthew J. Fogarty
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Wen-Zhi Zhan
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Vincent F. Simmon
- Spinogenix Inc, 4225 Executive Square, Suite 600 La Jolla, California, USA
| | | | - Stella T. Sarraf
- Spinogenix Inc, 4225 Executive Square, Suite 600 La Jolla, California, USA
| | - Gary C. Sieck
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
11
|
Han KA, Ko J. Orchestration of synaptic functions by WAVE regulatory complex-mediated actin reorganization. Exp Mol Med 2023; 55:1065-1075. [PMID: 37258575 PMCID: PMC10318009 DOI: 10.1038/s12276-023-01004-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/13/2023] [Accepted: 03/13/2023] [Indexed: 06/02/2023] Open
Abstract
The WAVE regulatory complex (WRC), composed of five components-Cyfip1/Sra1, WAVE/Scar, Abi, Nap1/Nckap1, and Brk1/HSPC300-is essential for proper actin cytoskeletal dynamics and remodeling in eukaryotic cells, likely by matching various patterned signals to Arp2/3-mediated actin nucleation. Accumulating evidence from recent studies has revealed diverse functions of the WRC in neurons, demonstrating its crucial role in dictating the assembly of molecular complexes for the patterning of various trans-synaptic signals. In this review, we discuss recent exciting findings on the physiological role of the WRC in regulating synaptic properties and highlight the involvement of WRC dysfunction in various brain disorders.
Collapse
Affiliation(s)
- Kyung Ah Han
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu, 42988, Korea
- Center for Synapse Diversity and Specificity, DGIST, Daegu, 42988, Korea
| | - Jaewon Ko
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu, 42988, Korea.
- Center for Synapse Diversity and Specificity, DGIST, Daegu, 42988, Korea.
| |
Collapse
|
12
|
Garner CC, Ackermann F. Synaptic logistics: The presynaptic scaffold protein Piccolo a nodal point tuning synaptic vesicle recycling, maintenance and integrity. Mol Cell Neurosci 2023; 124:103795. [PMID: 36436725 DOI: 10.1016/j.mcn.2022.103795] [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: 09/30/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
Properly working synapses are one important guarantor for a functional and healthy brain. They are small, densely packed structures, where information is transmitted through the release of neurotransmitters from synaptic vesicles (SVs). The latter cycle within the presynaptic terminal as they first fuse with the plasma membrane to deliver their neurotransmitter, and afterwards become recycled and prepared for a new release event. The synapse is an autonomous structure functioning mostly independent of the neuronal soma. Dysfunction in synaptic processes associated with local insults or genetic abnormalities can directly compromise synapse function and integrity and subsequently lead to the onset of neurodegenerative diseases. Therefore, measures need to be in place counteracting these threats for instance through the continuous replacement of old and damaged SV proteins. Interestingly recent studies show that the presynaptic scaffolding protein Piccolo contributes to health, function and integrity of synapses, as it mediates the delivery of synaptic proteins from the trans-Golgi network (TGN) towards synapses, as well as the local recycling and turnover of SV proteins within synaptic terminals. It can fulfill these various tasks through its multi-domain structure and ability to interact with numerous binding partners. In addition, Piccolo has recently been linked with the early onset neurodegenerative disease Pontocerebellar Hypoplasia Type 3 (PCH3) further underlying its importance for neuronal health. In this review, we will focus on Piccolo's contributions to synapse function, health and integrity and make a connection how those may contribute to the disease pattern of PCH3.
Collapse
Affiliation(s)
- Craig C Garner
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany; NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Germany
| | - Frauke Ackermann
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany.
| |
Collapse
|
13
|
Wu X, Qiu H, Zhang M. Interactions between Membraneless Condensates and Membranous Organelles at the Presynapse: A Phase Separation View of Synaptic Vesicle Cycle. J Mol Biol 2023; 435:167629. [PMID: 35595170 DOI: 10.1016/j.jmb.2022.167629] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 02/04/2023]
Abstract
Action potential-induced neurotransmitter release in presynaptic boutons involves coordinated actions of a large list of proteins that are associated directly or indirectly with membrane structures including synaptic vesicles and plasma membranes. These proteins are often highly abundant in different synaptic bouton sub-compartments, and they rarely act alone. Instead, these proteins interact with each other forming intricate and distinct molecular complexes. Many of these complexes form condensed clusters on membrane surfaces. This review summarizes findings in recent years showing that many of presynaptic protein complex assemblies are formed via phase separation. These protein condensates extensively interact with lipid membranes via distinct modes, forming various mesoscale structures by different mode of organizations between membraneless condensates and membranous organelles. We discuss that such mesoscale interactions could have deep implications on mobilization, exocytosis, and retrieval of synaptic vesicles.
Collapse
Affiliation(s)
- Xiandeng Wu
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Hua Qiu
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Mingjie Zhang
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen 518036, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
14
|
Nguyen NM, Vellichirammal NN, Guda C, Pendyala G. Decoding the Synaptic Proteome with Long-Term Exposure to Midazolam during Early Development. Int J Mol Sci 2022; 23:ijms23084137. [PMID: 35456952 PMCID: PMC9027542 DOI: 10.3390/ijms23084137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 12/05/2022] Open
Abstract
The intensive use of anesthetic and sedative agents in the neonatal intensive care unit (NICU) has raised controversial concerns about the potential neurodevelopmental risks. This study focused on midazolam (MDZ), a common benzodiazepine regularly used as a sedative on neonates in the NICU. Mounting evidence suggests a single exposure to MDZ during the neonatal period leads to learning disturbances. However, a knowledge gap that remains is how long-term exposure to MDZ during very early stages of life impacts synaptic alterations. Using a preclinical rodent model system, we mimicked a dose-escalation regimen on postnatal day 3 (P3) pups until day 21. Next, purified synaptosomes from P21 control and MDZ animals were subjected to quantitative mass-spectrometry-based proteomics, to identify potential proteomic signatures. Further analysis by ClueGO identified enrichment of proteins associated with actin-binding and protein depolymerization process. One potential hit identified was alpha adducin (ADD1), belonging to the family of cytoskeleton proteins, which was upregulated in the MDZ group and whose expression was further validated by Western blot. In summary, this study sheds new information on the long-term exposure of MDZ during the early stages of development impacts synaptic function, which could subsequently perturb neurobehavioral outcomes at later stages of life.
Collapse
Affiliation(s)
- Nghi M. Nguyen
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (N.N.V.); (C.G.)
| | - Neetha N. Vellichirammal
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (N.N.V.); (C.G.)
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (N.N.V.); (C.G.)
| | - Gurudutt Pendyala
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (N.N.V.); (C.G.)
- Child Health Research Institute, Omaha, NE 68198, USA
- Correspondence: ; Tel.: +1-402-559-8690
| |
Collapse
|
15
|
α-Synuclein at the Presynaptic Axon Terminal as a Double-Edged Sword. Biomolecules 2022; 12:biom12040507. [PMID: 35454096 PMCID: PMC9029495 DOI: 10.3390/biom12040507] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 12/12/2022] Open
Abstract
α-synuclein (α-syn) is a presynaptic, lipid-binding protein strongly associated with the neuropathology observed in Parkinson’s disease (PD), dementia with Lewy bodies (DLB), and Alzheimer’s Disease (AD). In normal physiology, α-syn plays a pivotal role in facilitating endocytosis and exocytosis. Interestingly, mutations and modifications of precise α-syn domains interfere with α-syn oligomerization and nucleation that negatively affect presynaptic vesicular dynamics, protein expressions, and mitochondrial profiles. Furthermore, the integration of the α-syn oligomers into the presynaptic membrane results in pore formations, ion influx, and excitotoxicity. Targeted therapies against specific domains of α-syn, including the use of small organic molecules, monoclonal antibodies, and synthetic peptides, are being screened and developed. However, the prospect of an effective α-syn targeted therapy is still plagued by low permeability across the blood–brain barrier (BBB), and poor entry into the presynaptic axon terminals. The present review proposes a modification of current strategies, which includes the use of novel encapsulation technology, such as lipid nanoparticles, to bypass the BBB and deliver such agents into the brain.
Collapse
|
16
|
Control of Synapse Structure and Function by Actin and Its Regulators. Cells 2022; 11:cells11040603. [PMID: 35203254 PMCID: PMC8869895 DOI: 10.3390/cells11040603] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/30/2022] [Accepted: 02/06/2022] [Indexed: 02/07/2023] Open
Abstract
Neurons transmit and receive information at specialized junctions called synapses. Excitatory synapses form at the junction between a presynaptic axon terminal and a postsynaptic dendritic spine. Supporting the shape and function of these junctions is a complex network of actin filaments and its regulators. Advances in microscopic techniques have enabled studies of the organization of actin at synapses and its dynamic regulation. In addition to highlighting recent advances in the field, we will provide a brief historical perspective of the understanding of synaptic actin at the synapse. We will also highlight key neuronal functions regulated by actin, including organization of proteins in the pre- and post- synaptic compartments and endocytosis of ion channels. We review the evidence that synapses contain distinct actin pools that differ in their localization and dynamic behaviors and discuss key functions for these actin pools. Finally, whole exome sequencing of humans with neurodevelopmental and psychiatric disorders has identified synaptic actin regulators as key disease risk genes. We briefly summarize how genetic variants in these genes impact neurotransmission via their impact on synaptic actin.
Collapse
|
17
|
Vallés AS, Barrantes FJ. Nanoscale Sub-Compartmentalization of the Dendritic Spine Compartment. Biomolecules 2021; 11:1697. [PMID: 34827695 PMCID: PMC8615865 DOI: 10.3390/biom11111697] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 01/04/2023] Open
Abstract
Compartmentalization of the membrane is essential for cells to perform highly specific tasks and spatially constrained biochemical functions in topographically defined areas. These membrane lateral heterogeneities range from nanoscopic dimensions, often involving only a few molecular constituents, to micron-sized mesoscopic domains resulting from the coalescence of nanodomains. Short-lived domains lasting for a few milliseconds coexist with more stable platforms lasting from minutes to days. This panoply of lateral domains subserves the great variety of demands of cell physiology, particularly high for those implicated in signaling. The dendritic spine, a subcellular structure of neurons at the receiving (postsynaptic) end of central nervous system excitatory synapses, exploits this compartmentalization principle. In its most frequent adult morphology, the mushroom-shaped spine harbors neurotransmitter receptors, enzymes, and scaffolding proteins tightly packed in a volume of a few femtoliters. In addition to constituting a mesoscopic lateral heterogeneity of the dendritic arborization, the dendritic spine postsynaptic membrane is further compartmentalized into spatially delimited nanodomains that execute separate functions in the synapse. This review discusses the functional relevance of compartmentalization and nanodomain organization in synaptic transmission and plasticity and exemplifies the importance of this parcelization in various neurotransmitter signaling systems operating at dendritic spines, using two fast ligand-gated ionotropic receptors, the nicotinic acetylcholine receptor and the glutamatergic receptor, and a second-messenger G-protein coupled receptor, the cannabinoid receptor, as paradigmatic examples.
Collapse
Affiliation(s)
- Ana Sofía Vallés
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (UNS-CONICET), Bahía Blanca 8000, Argentina;
| | - Francisco J. Barrantes
- Laboratory of Molecular Neurobiology, Institute of Biomedical Research (BIOMED), UCA-CONICET, Av. Alicia Moreau de Justo 1600, Buenos Aires C1107AFF, Argentina
| |
Collapse
|
18
|
Del Signore SJ, Kelley CF, Messelaar EM, Lemos T, Marchan MF, Ermanoska B, Mund M, Fai TG, Kaksonen M, Rodal AA. An autoinhibitory clamp of actin assembly constrains and directs synaptic endocytosis. eLife 2021; 10:69597. [PMID: 34324418 PMCID: PMC8321554 DOI: 10.7554/elife.69597] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/21/2021] [Indexed: 01/05/2023] Open
Abstract
Synaptic membrane-remodeling events such as endocytosis require force-generating actin assembly. The endocytic machinery that regulates these actin and membrane dynamics localizes at high concentrations to large areas of the presynaptic membrane, but actin assembly and productive endocytosis are far more restricted in space and time. Here we describe a mechanism whereby autoinhibition clamps the presynaptic endocytic machinery to limit actin assembly to discrete functional events. We found that collective interactions between the Drosophila endocytic proteins Nwk/FCHSD2, Dap160/intersectin, and WASp relieve Nwk autoinhibition and promote robust membrane-coupled actin assembly in vitro. Using automated particle tracking to quantify synaptic actin dynamics in vivo, we discovered that Nwk-Dap160 interactions constrain spurious assembly of WASp-dependent actin structures. These interactions also promote synaptic endocytosis, suggesting that autoinhibition both clamps and primes the synaptic endocytic machinery, thereby constraining actin assembly to drive productive membrane remodeling in response to physiological cues. Neurons constantly talk to each other by sending chemical signals across the tiny gap, or ‘synapse’, that separates two cells. While inside the emitting cell, these molecules are safely packaged into small, membrane-bound vessels. Upon the right signal, the vesicles fuse with the external membrane of the neuron and spill their contents outside, for the receiving cell to take up and decode. The emitting cell must then replenish its vesicle supply at the synapse through a recycling mechanism known as endocytosis. To do so, it uses dynamically assembling rod-like ‘actin’ filaments, which work in concert with many other proteins to pull in patches of membrane as new vesicles. The proteins that control endocytosis and actin assembly abound at neuronal synapses, and, when mutated, are linked to many neurological diseases. Unlike other cell types, neurons appear to ‘pre-deploy’ these actin-assembly proteins to synaptic membranes, but to keep them inactive under normal conditions. How neurons control the way this machinery is recruited and activated remains unknown. To investigate this question, Del Signore et al. conducted two sets of studies. First, they exposed actin to several different purified proteins in initial ‘test tube’ experiments. This revealed that, depending on the conditions, a group of endocytosis proteins could prevent or promote actin assembly: assembly occurred only if the proteins were associated with membranes. Next, Del Signore et al. mutated these proteins in fruit fly larvae, and performed live cell microscopy to determine their impact on actin assembly and endocytosis. Consistent with the test tube findings, endocytosis mutants had more actin assembly overall, implying that the proteins were required to prevent random actin assembly. However, the same mutants had reduced levels of endocytosis, suggesting that the proteins were also necessary for productive actin assembly. Together, these experiments suggest that, much like a mousetrap holds itself poised ready to spring, some endocytic proteins play a dual role to restrain actin assembly when and where it is not needed, and to promote it at sites of endocytosis. These results shed new light on how neurons might build and maintain effective, working synapses. Del Signore et al. hope that this knowledge may help to better understand and combat neurological diseases, such as Alzheimer’s, which are linked to impaired membrane traffic and cell signalling.
Collapse
Affiliation(s)
| | | | | | - Tania Lemos
- Department of Biology, Brandeis University, Walltham, United States
| | | | | | - Markus Mund
- Department of Biochemistry and NCCR Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Thomas G Fai
- Department of Mathematics, Brandeis University, Waltham, United States
| | - Marko Kaksonen
- Department of Biochemistry and NCCR Chemical Biology, University of Geneva, Geneva, Switzerland
| | | |
Collapse
|
19
|
Insulin and Leptin/Upd2 Exert Opposing Influences on Synapse Number in Fat-Sensing Neurons. Cell Metab 2020; 32:786-800.e7. [PMID: 32976758 PMCID: PMC7642105 DOI: 10.1016/j.cmet.2020.08.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 06/29/2020] [Accepted: 08/28/2020] [Indexed: 01/20/2023]
Abstract
Energy-sensing neural circuits decide to expend or conserve resources based, in part, on the tonic, steady-state, energy-store information they receive. Tonic signals, in the form of adipose tissue-derived adipokines, set the baseline level of activity in the energy-sensing neurons, thereby providing context for interpretation of additional inputs. However, the mechanism by which tonic adipokine information establishes steady-state neuronal function has heretofore been unclear. We show here that under conditions of nutrient surplus, Upd2, a Drosophila leptin ortholog, regulates actin-based synapse reorganization to reduce bouton number in an inhibitory circuit, thus establishing a neural tone that is permissive for insulin release. Unexpectedly, we found that insulin feeds back on these same inhibitory neurons to conversely increase bouton number, resulting in maintenance of negative tone. Our results point to a mechanism by which two surplus-sensing hormonal systems, Upd2/leptin and insulin, converge on a neuronal circuit with opposing outcomes to establish energy-store-dependent neuron activity.
Collapse
|
20
|
Abstract
It is increasingly recognized that local protein synthesis (LPS) contributes to fundamental aspects of axon biology, in both developing and mature neurons. Mutations in RNA-binding proteins (RBPs), as central players in LPS, and other proteins affecting RNA localization and translation are associated with a range of neurological disorders, suggesting disruption of LPS may be of pathological significance. In this review, we substantiate this hypothesis by examining the link between LPS and key axonal processes, and the implicated pathophysiological consequences of dysregulated LPS. First, we describe how the length and autonomy of axons result in an exceptional reliance on LPS. We next discuss the roles of LPS in maintaining axonal structural and functional polarity and axonal trafficking. We then consider how LPS facilitates the establishment of neuronal connectivity through regulation of axonal branching and pruning, how it mediates axonal survival into adulthood and its involvement in neuronal stress responses.
Collapse
Affiliation(s)
- Julie Qiaojin Lin
- UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Island Research Building, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Christine E Holt
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
21
|
Monday HR, Bourdenx M, Jordan BA, Castillo PE. CB 1-receptor-mediated inhibitory LTD triggers presynaptic remodeling via protein synthesis and ubiquitination. eLife 2020; 9:54812. [PMID: 32902378 PMCID: PMC7521925 DOI: 10.7554/elife.54812] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 09/08/2020] [Indexed: 01/03/2023] Open
Abstract
Long-lasting forms of postsynaptic plasticity commonly involve protein synthesis-dependent structural changes of dendritic spines. However, the relationship between protein synthesis and presynaptic structural plasticity remains unclear. Here, we investigated structural changes in cannabinoid-receptor 1 (CB1)-mediated long-term depression of inhibitory transmission (iLTD), a form of presynaptic plasticity that involves a protein-synthesis-dependent long-lasting reduction in GABA release. We found that CB1-iLTD in acute rat hippocampal slices was associated with protein synthesis-dependent presynaptic structural changes. Using proteomics, we determined that CB1 activation in hippocampal neurons resulted in increased ribosomal proteins and initiation factors, but decreased levels of proteins involved in regulation of the actin cytoskeleton, such as ARPC2 and WASF1/WAVE1, and presynaptic release. Moreover, while CB1-iLTD increased ubiquitin/proteasome activity, ubiquitination but not proteasomal degradation was critical for structural and functional presynaptic CB1-iLTD. Thus, CB1-iLTD relies on both protein synthesis and ubiquitination to elicit structural changes that underlie long-term reduction of GABA release.
Collapse
Affiliation(s)
- Hannah R Monday
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, United States
| | - Mathieu Bourdenx
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, United States.,Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, United States
| | - Bryen A Jordan
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, United States.,Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, United States
| | - Pablo E Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, United States.,Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, United States
| |
Collapse
|
22
|
McNeill EM, Thompson C, Berke B, Chou VT, Rusch J, Duckworth A, DeProto J, Taylor A, Gates J, Gertler F, Keshishian H, Van Vactor D. Drosophila enabled promotes synapse morphogenesis and regulates active zone form and function. Neural Dev 2020; 15:4. [PMID: 32183907 PMCID: PMC7076993 DOI: 10.1186/s13064-020-00141-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 02/25/2020] [Indexed: 11/10/2022] Open
Abstract
Background Recent studies of synapse form and function highlight the importance of the actin cytoskeleton in regulating multiple aspects of morphogenesis, neurotransmission, and neural plasticity. The conserved actin-associated protein Enabled (Ena) is known to regulate development of the Drosophila larval neuromuscular junction through a postsynaptic mechanism. However, the functions and regulation of Ena within the presynaptic terminal has not been determined. Methods Here, we use a conditional genetic approach to address a presynaptic role for Ena on presynaptic morphology and ultrastructure, and also examine the pathway in which Ena functions through epistasis experiments. Results We find that Ena is required to promote the morphogenesis of presynaptic boutons and branches, in contrast to its inhibitory role in muscle. Moreover, while postsynaptic Ena is regulated by microRNA-mediated mechanisms, presynaptic Ena relays the output of the highly conserved receptor protein tyrosine phosphatase Dlar and associated proteins including the heparan sulfate proteoglycan Syndecan, and the non-receptor Abelson tyrosine kinase to regulate addition of presynaptic varicosities. Interestingly, Ena also influences active zones, where it restricts active zone size, regulates the recruitment of synaptic vesicles, and controls the amplitude and frequency of spontaneous glutamate release. Conclusion We thus show that Ena, under control of the Dlar pathway, is required for presynaptic terminal morphogenesis and bouton addition and that Ena has active zone and neurotransmission phenotypes. Notably, in contrast to Dlar, Ena appears to integrate multiple pathways that regulate synapse form and function.
Collapse
Affiliation(s)
- Elizabeth M McNeill
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA.
| | - Cheryl Thompson
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Brett Berke
- Department of Biology, Yale University, New Haven, CT, USA
| | - Vivian T Chou
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| | - Jannette Rusch
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - April Duckworth
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Jamin DeProto
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Alicia Taylor
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA.,Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Julie Gates
- Department of Biology, Bucknell University, Lewisburg, PA, USA
| | - Frank Gertler
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, England
| | | | - David Van Vactor
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
23
|
Yang S, Zhao Y, Chen X, Lu X, Chen Y, Zhao X, Zhu L, Fang Z, Zhao H, Yao Y, Liu C, Shen C. The ACTB Variants and Alcohol Drinking Confer Joint Effect to Ischemic Stroke in Chinese Han Population. J Atheroscler Thromb 2020; 27:226-244. [PMID: 31327802 PMCID: PMC7113141 DOI: 10.5551/jat.49536] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 05/28/2019] [Indexed: 12/11/2022] Open
Abstract
AIM β-actin (ACTB) participates in the vascular remodeling and contributes to the cardiovascular diseases. Herein, we investigated the associations of ACTB with hypertension and stroke. METHODS Three single-nucleotide polymorphisms in ACTB were selected for genotyping in 2,012 hypertension cases and 2,210 controls. The associations of ACTB with hypertension and stroke were examined in another follow-up study. Logistic and Cox regression were performed in a case-control study and a follow-up study, respectively. Additive scale interaction was examined by calculating the relative excess risk due to interaction (RERI), attributable proportion due to interaction (AP) and synergy index (SI). The multiplicative interaction hazard ratio was calculated by fitting the Cox regression model. ACTB mRNA in peripheral blood mononuclear cells was measured in ischemic stroke (IS) cases and in controls. RESULTS The associations of rs852426 with hypertension and stroke had statistical significance in drinkers but not after Bonferroni correction. An additive interaction of rs852426 and drinking was observed for stroke incidence, the adjusted RERI was -0.907 (p=4.108×10-4), and the multiplicative interaction was still sound (HR=0.541, p=0.048). Furthermore, the significant interaction was further replicated in a nested case-control study. In the drinking population, the relative expression of ACTB mRNA in IS was lower (0.99±0.26) than that in controls (1.13±0.20), with a p value of 0.026. CONCLUSIONS ACTB rs852426 was significantly associated with alcohol consumption on stroke risk, and the expression of ACTB mRNA in IS who had a drinking habit was significantly down-regulated. This finding will provide a novel insight into the prevention of stroke.
Collapse
Affiliation(s)
- Song Yang
- Department of Cardiology, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, China
| | - Yanping Zhao
- Department of Neurology, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, China
| | - Xiaotian Chen
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiangfeng Lu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanchun Chen
- Department of Cardiology, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, China
| | - Xianghai Zhao
- Department of Cardiology, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, China
| | - Lijun Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College, Wuhu, China
| | - Zhengmei Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College, Wuhu, China
| | - Hailong Zhao
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yingshui Yao
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College, Wuhu, China
| | - Chunlan Liu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chong Shen
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
24
|
Eshraghi M, Gombar R, De Repentigny Y, Vacratsis PO, Kothary R. Pathologic Alterations in the Proteome of Synaptosomes from a Mouse Model of Spinal Muscular Atrophy. J Proteome Res 2019; 18:3042-3051. [PMID: 31262178 DOI: 10.1021/acs.jproteome.9b00159] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Spinal muscular atrophy (SMA) is a human genetic disorder characterized by muscle weakness, muscle atrophy, and death of motor neurons. SMA is caused by mutations or deletions in a gene called survival motor neuron 1 (SMN1). SMN1 is a housekeeping gene, but the most prominent pathologies in SMA are atrophy of myofibers and death of motor neurons. Further, degeneration of neuromuscular junctions, of synapses, and of axonal regions are features of SMA disease. Here, we have investigated the proteome dynamics of central synapses in P14 Smn2B/- mice, a model of SMA. Label-free quantitative proteomics on isolated synaptosomes from spinal cords of these animals identified 2030 protein groups. Statistical data analysis revealed 65 specific alterations in the proteome of the central synapses at the early onset stage of disease. Functional analysis of the dysregulated proteins indicated a significant enrichment of proteins associated with mitochondrial dynamics, cholesterol biogenesis, and protein clearance. These pathways represent potential targets for therapy development with the goal of providing stability to the central synapses, thereby preserving neuronal integrity in the context of SMA disease. Data are available via ProteomeXchange with identifier PXD012850.
Collapse
Affiliation(s)
- Mehdi Eshraghi
- Regenerative Medicine Program , Ottawa Hospital Research Institute , Ottawa , Ontario K1H 8L6 , Canada.,University of Ottawa Centre for Neuromuscular Disease , Ottawa , Ontario K1H 8M5 , Canada
| | - Robert Gombar
- Department of Chemistry and Biochemistry , University of Windsor , Windsor , Ontario N9B 3P4 , Canada
| | - Yves De Repentigny
- Regenerative Medicine Program , Ottawa Hospital Research Institute , Ottawa , Ontario K1H 8L6 , Canada
| | - Panayiotis O Vacratsis
- Department of Chemistry and Biochemistry , University of Windsor , Windsor , Ontario N9B 3P4 , Canada
| | - Rashmi Kothary
- Regenerative Medicine Program , Ottawa Hospital Research Institute , Ottawa , Ontario K1H 8L6 , Canada.,University of Ottawa Centre for Neuromuscular Disease , Ottawa , Ontario K1H 8M5 , Canada
| |
Collapse
|
25
|
Anbalagan S, Blechman J, Gliksberg M, Gordon L, Rotkopf R, Dadosh T, Shimoni E, Levkowitz G. Robo2 regulates synaptic oxytocin content by affecting actin dynamics. eLife 2019; 8:45650. [PMID: 31180321 PMCID: PMC6590984 DOI: 10.7554/elife.45650] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 06/08/2019] [Indexed: 12/28/2022] Open
Abstract
The regulation of neuropeptide level at the site of release is essential for proper neurophysiological functions. We focused on a prominent neuropeptide, oxytocin (OXT) in the zebrafish as an in vivo model to visualize and quantify OXT content at the resolution of a single synapse. We found that OXT-loaded synapses were enriched with polymerized actin. Perturbation of actin filaments by either cytochalasin-D or conditional Cofilin expression resulted in decreased synaptic OXT levels. Genetic loss of robo2 or slit3 displayed decreased synaptic OXT content and robo2 mutants displayed reduced mobility of the actin probe Lifeact-EGFP in OXT synapses. Using a novel transgenic reporter allowing real-time monitoring of OXT-loaded vesicles, we show that robo2 mutants display slower rate of vesicles accumulation. OXT-specific expression of dominant-negative Cdc42, which is a key regulator of actin dynamics and a downstream effector of Robo2, led to a dose-dependent increase in OXT content in WT, and a dampened effect in robo2 mutants. Our results link Slit3-Robo2-Cdc42, which controls local actin dynamics, with the maintenance of synaptic neuropeptide levels.
Collapse
Affiliation(s)
- Savani Anbalagan
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Janna Blechman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Michael Gliksberg
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ludmila Gordon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ron Rotkopf
- Bioinformatics Unit, LSCF, Weizmann Institute of Science, Rehovot, Israel.,Electron Microscopy Unit, Weizmann Institute of Science, Rehovot, Israel
| | - Tali Dadosh
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal Shimoni
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Gil Levkowitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
26
|
Guzman GA, Guzman RE, Jordan N, Hidalgo P. A Tripartite Interaction Among the Calcium Channel α 1- and β-Subunits and F-Actin Increases the Readily Releasable Pool of Vesicles and Its Recovery After Depletion. Front Cell Neurosci 2019; 13:125. [PMID: 31130843 PMCID: PMC6509170 DOI: 10.3389/fncel.2019.00125] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 03/13/2019] [Indexed: 11/13/2022] Open
Abstract
Neurotransmitter release is initiated by the influx of Ca2+via voltage-gated calcium channels. The accessory β-subunit (CaVβ) of these channels shapes synaptic transmission by associating with the pore-forming subunit (CaVα1) and up-regulating presynaptic calcium currents. Besides CaVα1, CaVβ interacts with several partners including actin filaments (F-actin). These filaments are known to associate with synaptic vesicles (SVs) at the presynaptic terminals and support their translocation within different pools, but the role of CaVβ/F-actin association on synaptic transmission has not yet been explored. We here study how CaVβ4, the major calcium channel β isoform in mamalian brain, modifies synaptic transmission in concert with F-actin in cultured hippocampal neurons. We analyzed the effect of exogenous CaVβ4 before and after pharmacological disruption of the actin cytoskeleton and dissected calcium channel-dependent and -independent functions by comparing the effects of the wild-type subunit with the one bearing a double mutation that impairs binding to CaVα1. We found that exogenously expressed wild-type CaVβ4 enhances spontaneous and depolarization-evoked excitatory postsynaptic currents (EPSCs) without altering synaptogenesis. CaVβ4 increases the size of the readily releasable pool (RRP) of SVs at resting conditions and accelerates their recovery after depletion. The enhanced neurotransmitter release induced by CaVβ4 is abolished upon disruption of the actin cytoskeleton. The CaVα1 association-deficient CaVβ4 mutant associates with actin filaments, but neither alters postsynaptic responses nor the time course of the RRP recovery. Furthermore, this mutant protein preserves the ability to increase the RRP size. These results indicate that the interplay between CaVβ4 and F-actin also support the recruitment of SVs to the RRP in a CaVα1-independent manner. Our studies show an emerging role of CaVβ in determining SV maturation toward the priming state and its replenishment after release. We envision that this subunit plays a role in coupling exocytosis to endocytosis during the vesicle cycle.
Collapse
Affiliation(s)
- Gustavo A Guzman
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, Jülich, Germany
| | - Raul E Guzman
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, Jülich, Germany
| | - Nadine Jordan
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, Jülich, Germany
| | - Patricia Hidalgo
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, Jülich, Germany.,Institute of Biochemistry, Heinrich-Heine University, Düsseldorf, Germany
| |
Collapse
|
27
|
Rampérez A, Bartolomé-Martín D, García-Pascual A, Sánchez-Prieto J, Torres M. Photoconversion of FM1-43 Reveals Differences in Synaptic Vesicle Recycling and Sensitivity to Pharmacological Disruption of Actin Dynamics in Individual Synapses. ACS Chem Neurosci 2019; 10:2045-2059. [PMID: 30763065 DOI: 10.1021/acschemneuro.8b00712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The cycling of synaptic vesicles ensures that neurons can communicate adequately through their synapses on repeated occasions when activity is sustained, and several steps in this cycle are modulated by actin. The effects of pharmacological stabilization of actin with jasplakinolide or its depolymerization with latrunculin A was assessed on the synaptic vesicle cycle at individual boutons of cerebellar granule cells, using FM1-43 imaging to track vesicle recycling and its photoconversion to specifically label recycled organelles. Remarkable differences in the recycling capacity of individual boutons are evident, and their dependence on the actin cytoskeleton for recycling is clear. Disrupting actin dynamics causes a loss of functional boutons, and while this indicates that exo/endocytotic cycling in boutons is fully dependent on such events, this dependence is only partial in other boutons. Indeed, exocytosis and vesicle trafficking are impaired significantly by stabilizing or depolymerizing actin, whereas repositioning recycled vesicles at the active zone seems to be dependent on actin polymerization alone. These findings support the hypothesis that different steps of synaptic vesicle cycling depend on actin dynamics and that such dependence varies among individual boutons.
Collapse
Affiliation(s)
- Alberto Rampérez
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain
| | - David Bartolomé-Martín
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain
| | - Angeles García-Pascual
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain
| | - Jose Sánchez-Prieto
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain
| | - Magdalena Torres
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain
| |
Collapse
|
28
|
The role of LRRK2 in cytoskeletal dynamics. Biochem Soc Trans 2018; 46:1653-1663. [PMID: 30467120 DOI: 10.1042/bst20180469] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/16/2018] [Accepted: 10/24/2018] [Indexed: 12/11/2022]
Abstract
Leucine-rich repeat kinase 2 (LRRK2), a complex kinase/GTPase mutated in Parkinson's disease, has been shown to physically and functionally interact with cytoskeletal-related components in different brain cells. Neurons greatly rely on a functional cytoskeleton for many homeostatic processes such as local and long-distance vesicle transport, synaptic plasticity, and dendrites/axons growth and remodeling. Here, we will review the available data linking LRRK2 and the cytoskeleton, and discuss how this may be functionally relevant for the well-established roles of LRRK2 in intracellular trafficking pathways and outgrowth of neuronal processes in health and disease conditions.
Collapse
|
29
|
Lahbib S, Leblond CS, Hamza M, Regnault B, Lemée L, Mathieu A, Jaouadi H, Mkaouar R, Youssef-Turki IB, Belhadj A, Kraoua I, Bourgeron T, Abdelhak S. Homozygous 2p11.2 deletion supports the implication of ELMOD3 in hearing loss and reveals the potential association of CAPG with ASD/ID etiology. J Appl Genet 2018; 60:49-56. [PMID: 30284680 DOI: 10.1007/s13353-018-0472-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/25/2018] [Accepted: 09/27/2018] [Indexed: 11/30/2022]
Abstract
Autism spectrum disorder (ASD) is a set of neurodevelopmental conditions characterized by early-onset difficulties in social communication and unusually restricted, repetitive behavior and interests. Parental consanguinity may lead to higher risk of ASD and to more severe clinical presentations in the offspring. Studies of ASD families with high inbreeding enable the identification of inherited variants of this disorder particularly those with an autosomal recessive pattern of inheritance. In our study, using copy number variants (CNV) analysis, we identified a rare homozygous deletion in 2p11.2 region that affects ELMOD3, CAPG, and SH2D6 genes in a boy with ASD, intellectual disability (ID), and hearing impairment (HI). This deletion may reveal a new contiguous deletion syndrome in which ELMOD3, known to be implicated in autosomal recessive deafness underlies the HI of the proband and CAPG, member of actin regulatory proteins involved in cytoskeletal dynamic, an important function for brain development and activity, underlies the ASD/ID phenotype. A possible contribution of SH2D6 gene, as a part of a chimeric gene, to the clinical presentation of the patient is discussed. Our result supports the implication of ELMOD3 in hearing loss and highlights the potential clinical relevance of 2p11.2 deletion in autism and/or intellectual disability.
Collapse
Affiliation(s)
- Saida Lahbib
- Biomedical Genomics and Oncogenetics Laboratory LR16IPT05, Université Tunis El Manar, Institut Pasteur de Tunis, 1002, Tunis, Tunisia. .,University of Tunis El Manar, Tunis, Tunisia.
| | - Claire S Leblond
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, 75015, Paris, France.,CNRS UMR3571, Genes, Synapses and Cognition, Institut Pasteur, 75015, Paris, France.,Paris Diderot University, Sorbonne Paris Cité, 75013, Paris, France
| | - Mariem Hamza
- Faculty of Medicine of Tunis, University of Tunis El Manar, 1007 La Rabta, Tunis, Tunisia.,Child and Adolescent Psychiatry Department, Mongi Slim Hospital, 2046, Sidi Daoud, Tunisia
| | - Béatrice Regnault
- Plateforme de Génotypage des Eucaryotes, Centre d'Innovation et Recherche Technologique (CITECH), Institut Pasteur, 75015, Paris, France
| | - Laure Lemée
- Plateforme de Génotypage des Eucaryotes, Centre d'Innovation et Recherche Technologique (CITECH), Institut Pasteur, 75015, Paris, France
| | - Alexandre Mathieu
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, 75015, Paris, France.,CNRS UMR3571, Genes, Synapses and Cognition, Institut Pasteur, 75015, Paris, France.,Paris Diderot University, Sorbonne Paris Cité, 75013, Paris, France
| | - Hager Jaouadi
- Biomedical Genomics and Oncogenetics Laboratory LR16IPT05, Université Tunis El Manar, Institut Pasteur de Tunis, 1002, Tunis, Tunisia
| | - Rahma Mkaouar
- Biomedical Genomics and Oncogenetics Laboratory LR16IPT05, Université Tunis El Manar, Institut Pasteur de Tunis, 1002, Tunis, Tunisia
| | - Ilhem Ben Youssef-Turki
- Faculty of Medicine of Tunis, University of Tunis El Manar, 1007 La Rabta, Tunis, Tunisia.,Research Unit UR12 SP24 and Department of Child and Adolescent Neurology, National Institute Mongi Ben Hmida of Neurology, 1007, Tunis, Tunisia
| | - Ahlem Belhadj
- Faculty of Medicine of Tunis, University of Tunis El Manar, 1007 La Rabta, Tunis, Tunisia.,Child and Adolescent Psychiatry Department, Mongi Slim Hospital, 2046, Sidi Daoud, Tunisia
| | - Ichraf Kraoua
- Faculty of Medicine of Tunis, University of Tunis El Manar, 1007 La Rabta, Tunis, Tunisia.,Research Unit UR12 SP24 and Department of Child and Adolescent Neurology, National Institute Mongi Ben Hmida of Neurology, 1007, Tunis, Tunisia
| | - Thomas Bourgeron
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, 75015, Paris, France.,CNRS UMR3571, Genes, Synapses and Cognition, Institut Pasteur, 75015, Paris, France.,Paris Diderot University, Sorbonne Paris Cité, 75013, Paris, France
| | - Sonia Abdelhak
- Biomedical Genomics and Oncogenetics Laboratory LR16IPT05, Université Tunis El Manar, Institut Pasteur de Tunis, 1002, Tunis, Tunisia
| |
Collapse
|
30
|
Role of Actin Filament on Synaptic Vesicle Pooling in Cultured Hippocampal Neuron. Appl Microsc 2018. [DOI: 10.9729/am.2018.48.3.55] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
31
|
Joensuu M, Lanoue V, Hotulainen P. Dendritic spine actin cytoskeleton in autism spectrum disorder. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:362-381. [PMID: 28870634 DOI: 10.1016/j.pnpbp.2017.08.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/21/2017] [Accepted: 08/30/2017] [Indexed: 01/01/2023]
Abstract
Dendritic spines are small actin-rich protrusions from neuronal dendrites that form the postsynaptic part of most excitatory synapses. Changes in the shape and size of dendritic spines correlate with the functional changes in excitatory synapses and are heavily dependent on the remodeling of the underlying actin cytoskeleton. Recent evidence implicates synapses at dendritic spines as important substrates of pathogenesis in neuropsychiatric disorders, including autism spectrum disorder (ASD). Although synaptic perturbations are not the only alterations relevant for these diseases, understanding the molecular underpinnings of the spine and synapse pathology may provide insight into their etiologies and could reveal new drug targets. In this review, we will discuss recent findings of defective actin regulation in dendritic spines associated with ASD.
Collapse
Affiliation(s)
- Merja Joensuu
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland; Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Vanessa Lanoue
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Pirta Hotulainen
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland.
| |
Collapse
|
32
|
Migh E, Götz T, Földi I, Szikora S, Gombos R, Darula Z, Medzihradszky KF, Maléth J, Hegyi P, Sigrist S, Mihály J. Microtubule organization in presynaptic boutons relies on the formin DAAM. Development 2018; 145:dev158519. [PMID: 29487108 DOI: 10.1242/dev.158519] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 02/14/2018] [Indexed: 02/02/2023]
Abstract
Regulation of the cytoskeleton is fundamental to the development and function of synaptic terminals, such as neuromuscular junctions. Despite the identification of numerous proteins that regulate synaptic actin and microtubule dynamics, the mechanisms of cytoskeletal control during terminal arbor formation have remained largely elusive. Here, we show that DAAM, a member of the formin family of cytoskeleton organizing factors, is an important presynaptic regulator of neuromuscular junction development in Drosophila We demonstrate that the actin filament assembly activity of DAAM plays a negligible role in terminal formation; rather, DAAM is necessary for synaptic microtubule organization. Genetic interaction studies consistently link DAAM with the Wg/Ank2/Futsch module of microtubule regulation and bouton formation. Finally, we provide evidence that DAAM is tightly associated with the synaptic active zone scaffold, and electrophysiological data point to a role in the modulation of synaptic vesicle release. Based on these results, we propose that DAAM is an important cytoskeletal effector element of the Wg/Ank2 pathway involved in the determination of basic synaptic structures, and, additionally, that DAAM may couple the active zone scaffold to the presynaptic cytoskeleton.
Collapse
Affiliation(s)
- Ede Migh
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Torsten Götz
- Institut für Biologie/Genetik and NeuroCure, Freie Universitat Berlin, Takustrasse 6, D-14195 Berlin, Germany
| | - István Földi
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Szilárd Szikora
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Rita Gombos
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Zsuzsanna Darula
- Laboratory of Proteomics Research, Biological Research Centre, Hungarian Academy of Sciences, Szeged H-6726, Hungary
| | - Katalin F Medzihradszky
- Laboratory of Proteomics Research, Biological Research Centre, Hungarian Academy of Sciences, Szeged H-6726, Hungary
| | - József Maléth
- MTA-SZTE Translational Gastroenterology Research Group, Szeged H-6725, Hungary
| | - Péter Hegyi
- MTA-SZTE Translational Gastroenterology Research Group, Szeged H-6725, Hungary
- Institute for Translational Medicine, University of Pecs, Pécs H-7624, Hungary
| | - Stephan Sigrist
- Institut für Biologie/Genetik and NeuroCure, Freie Universitat Berlin, Takustrasse 6, D-14195 Berlin, Germany
| | - József Mihály
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| |
Collapse
|
33
|
Calahorro F, Izquierdo PG. The presynaptic machinery at the synapse of C. elegans. INVERTEBRATE NEUROSCIENCE : IN 2018; 18:4. [PMID: 29532181 PMCID: PMC5851683 DOI: 10.1007/s10158-018-0207-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 02/22/2018] [Indexed: 11/17/2022]
Abstract
Synapses are specialized contact sites that mediate information flow between neurons and their targets. Important physical interactions across the synapse are mediated by synaptic adhesion molecules. These adhesions regulate formation of synapses during development and play a role during mature synaptic function. Importantly, genes regulating synaptogenesis and axon regeneration are conserved across the animal phyla. Genetic screens in the nematode Caenorhabditis elegans have identified a number of molecules required for synapse patterning and assembly. C. elegans is able to survive even with its neuronal function severely compromised. This is in comparison with Drosophila and mice where increased complexity makes them less tolerant to impaired function. Although this fact may reflect differences in the function of the homologous proteins in the synapses between these organisms, the most likely interpretation is that many of these components are equally important, but not absolutely essential, for synaptic transmission to support the relatively undemanding life style of laboratory maintained C. elegans. Here, we review research on the major group of synaptic proteins, involved in the presynaptic machinery in C. elegans, showing a strong conservation between higher organisms and highlight how C. elegans can be used as an informative tool for dissecting synaptic components, based on a simple nervous system organization.
Collapse
Affiliation(s)
- Fernando Calahorro
- Biological Sciences, University of Southampton, Life Sciences Building 85, Southampton, SO17 1BJ, UK.
| | - Patricia G Izquierdo
- Biological Sciences, University of Southampton, Life Sciences Building 85, Southampton, SO17 1BJ, UK
| |
Collapse
|
34
|
Drosophila Syd-1 Has RhoGAP Activity That Is Required for Presynaptic Clustering of Bruchpilot/ELKS but Not Neurexin-1. Genetics 2017; 208:705-716. [PMID: 29217522 DOI: 10.1534/genetics.117.300538] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 11/22/2017] [Indexed: 12/23/2022] Open
Abstract
Syd-1 proteins are required for presynaptic development in worm, fly, and mouse. Syd-1 proteins in all three species contain a Rho GTPase activating protein (GAP)-like domain of unclear significance: invertebrate Syd-1s are thought to lack GAP activity, and mouse mSYD1A has GAP activity that is thought to be dispensable for its function. Here, we show that Drosophila melanogaster Syd-1 can interact with all six fly Rhos and has GAP activity toward Rac1 and Cdc42. During development, fly Syd-1 clusters multiple presynaptic proteins at the neuromuscular junction (NMJ), including the cell adhesion molecule Neurexin (Nrx-1) and the active zone (AZ) component Bruchpilot (Brp), both of which Syd-1 binds directly. We show that a mutant form of Syd-1 that specifically lacks GAP activity localizes normally to presynaptic sites and is sufficient to recruit Nrx-1 but fails to cluster Brp normally. We provide evidence that Syd-1 participates with Rac1 in two separate functions: (1) together with the Rac guanine exchange factor (RacGEF) Trio, GAP-active Syd-1 is required to regulate the nucleotide-bound state of Rac1, thereby promoting Brp clustering; and (2) Syd-1, independent of its GAP activity, is required for the recruitment of Nrx-1 to boutons, including the recruitment of Nrx-1 that is promoted by GTP-bound Rac1. We conclude that, contrary to current models, the GAP domain of fly Syd-1 is active and required for presynaptic development; we suggest that the same may be true of vertebrate Syd-1 proteins. In addition, our data provide new molecular insight into the ability of Rac1 to promote presynaptic development.
Collapse
|
35
|
Rui M, Qian J, Liu L, Cai Y, Lv H, Han J, Jia Z, Xie W. The neuronal protein Neurexin directly interacts with the Scribble-Pix complex to stimulate F-actin assembly for synaptic vesicle clustering. J Biol Chem 2017; 292:14334-14348. [PMID: 28710284 PMCID: PMC5582829 DOI: 10.1074/jbc.m117.794040] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 06/29/2017] [Indexed: 01/17/2023] Open
Abstract
Synaptic vesicles (SVs) form distinct pools at synaptic terminals, and this well-regulated separation is necessary for normal neurotransmission. However, how the SV cluster, in particular synaptic compartments, maintains normal neurotransmitter release remains a mystery. The presynaptic protein Neurexin (NRX) plays a significant role in synaptic architecture and function, and some evidence suggests that NRX is associated with neurological disorders, including autism spectrum disorders. However, the role of NRX in SV clustering is unclear. Here, using the neuromuscular junction at the 2-3 instar stages of Drosophila larvae as a model and biochemical imaging and electrophysiology techniques, we demonstrate that Drosophila NRX (DNRX) plays critical roles in regulating synaptic terminal clustering and release of SVs. We found that DNRX controls the terminal clustering and release of SVs by stimulating presynaptic F-actin. Furthermore, our results indicate that DNRX functions through the scaffold protein Scribble and the GEF protein DPix to activate the small GTPase Ras-related C3 Botulinum toxin substrate 1 (Rac1). We observed a direct interaction between the C-terminal PDZ-binding motif of DNRX and the PDZ domains of Scribble and that Scribble bridges DNRX to DPix, forming a DNRX-Scribble-DPix complex that activates Rac1 and subsequently stimulates presynaptic F-actin assembly and SV clustering. Taken together, our work provides important insights into the function of DNRX in regulating SV clustering, which could help inform further research into pathological neurexin-mediated mechanisms in neurological disorders such as autism.
Collapse
Affiliation(s)
- Menglong Rui
- From Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Jinjun Qian
- From Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Lijuan Liu
- From Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Yihan Cai
- From Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Huihui Lv
- From Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Junhai Han
- From Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China.,the Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing 210096, China
| | - Zhengping Jia
- the Department of Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada, and.,the Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Wei Xie
- From Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China, .,the Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing 210096, China
| |
Collapse
|
36
|
Torres VI, Inestrosa NC. Vertebrate Presynaptic Active Zone Assembly: a Role Accomplished by Diverse Molecular and Cellular Mechanisms. Mol Neurobiol 2017; 55:4513-4528. [PMID: 28685386 DOI: 10.1007/s12035-017-0661-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/14/2017] [Indexed: 01/22/2023]
Abstract
Among all the biological systems in vertebrates, the central nervous system (CNS) is the most complex, and its function depends on specialized contacts among neurons called synapses. The assembly and organization of synapses must be exquisitely regulated for a normal brain function and network activity. There has been a tremendous effort in recent decades to understand the molecular and cellular mechanisms participating in the formation of new synapses and their organization, maintenance, and regulation. At the vertebrate presynapses, proteins such as Piccolo, Bassoon, RIM, RIM-BPs, CAST/ELKS, liprin-α, and Munc13 are constant residents and participate in multiple and dynamic interactions with other regulatory proteins, which define network activity and normal brain function. Here, we review the function of these active zone (AZ) proteins and diverse factors involved in AZ assembly and maintenance, with an emphasis on axonal trafficking of precursor vesicles, protein homo- and hetero-oligomeric interactions as a mechanism of AZ trapping and stabilization, and the role of F-actin in presynaptic assembly and its modulation by Wnt signaling.
Collapse
Affiliation(s)
- Viviana I Torres
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Center for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia. .,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.
| |
Collapse
|
37
|
Ross JA, Webster RG, Lechertier T, Reynolds LE, Turmaine M, Bencze M, Jamshidi Y, Cetin H, Muntoni F, Beeson D, Hodilvala-Dilke K, Conti FJ. Multiple roles of integrin-α3 at the neuromuscular junction. J Cell Sci 2017; 130:1772-1784. [PMID: 28386022 PMCID: PMC5450193 DOI: 10.1242/jcs.201103] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 03/31/2017] [Indexed: 12/22/2022] Open
Abstract
The neuromuscular junction (NMJ) is the synapse between motoneurons and skeletal muscle, and is responsible for eliciting muscle contraction. Neurotransmission at synapses depends on the release of synaptic vesicles at sites called active zones (AZs). Various proteins of the extracellular matrix are crucial for NMJ development; however, little is known about the identity and functions of the receptors that mediate their effects. Using genetically modified mice, we find that integrin-α3 (encoded by Itga3), an adhesion receptor at the presynaptic membrane, is involved in the localisation of AZ components and efficient synaptic vesicle release. Integrin-α3 also regulates integrity of the synapse - mutant NMJs present with progressive structural changes and upregulated autophagy, features commonly observed during ageing and in models of neurodegeneration. Unexpectedly, we find instances of nerve terminal detachment from the muscle fibre; to our knowledge, this is the first report of a receptor that is required for the physical anchorage of pre- and postsynaptic elements at the NMJ. These results demonstrate multiple roles of integrin-α3 at the NMJ, and suggest that alterations in its function could underlie defects that occur in neurodegeneration or ageing.
Collapse
Affiliation(s)
- Jacob A Ross
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Richard G Webster
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Tanguy Lechertier
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Louise E Reynolds
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Mark Turmaine
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Maximilien Bencze
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Yalda Jamshidi
- Department of Genetics, Institute of Molecular and Clinical Sciences, St George's University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Hakan Cetin
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - David Beeson
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Kairbaan Hodilvala-Dilke
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Francesco J Conti
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
38
|
Mallik B, Dwivedi MK, Mushtaq Z, Kumari M, Verma PK, Kumar V. Regulation of neuromuscular junction organization by Rab2 and its effector ICA69 in Drosophila. Development 2017; 144:2032-2044. [PMID: 28455372 DOI: 10.1242/dev.145920] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 04/19/2017] [Indexed: 12/31/2022]
Abstract
The mechanisms underlying synaptic differentiation, which involves neuronal membrane and cytoskeletal remodeling, are not completely understood. We performed a targeted RNAi-mediated screen of Drosophila BAR-domain proteins and identified islet cell autoantigen 69 kDa (ICA69) as one of the key regulators of morphological differentiation of the larval neuromuscular junction (NMJ). We show that Drosophila ICA69 colocalizes with α-Spectrin at the NMJ. The conserved N-BAR domain of ICA69 deforms liposomes in vitro Full-length ICA69 and the ICAC but not the N-BAR domain of ICA69 induce filopodia in cultured cells. Consistent with its cytoskeleton regulatory role, ICA69 mutants show reduced α-Spectrin immunoreactivity at the larval NMJ. Manipulating levels of ICA69 or its interactor PICK1 alters the synaptic level of ionotropic glutamate receptors (iGluRs). Moreover, reducing PICK1 or Rab2 levels phenocopies ICA69 mutation. Interestingly, Rab2 regulates not only synaptic iGluR but also ICA69 levels. Thus, our data suggest that: (1) ICA69 regulates NMJ organization through a pathway that involves PICK1 and Rab2, and (2) Rab2 functions genetically upstream of ICA69 and regulates NMJ organization and targeting/retention of iGluRs by regulating ICA69 levels.
Collapse
Affiliation(s)
- Bhagaban Mallik
- Department of Biological Sciences, AB-3, Indian Institute of Science Education and Research, Bhauri, Bhopal, Madhya Pradesh 462066, India
| | - Manish Kumar Dwivedi
- Department of Biological Sciences, AB-3, Indian Institute of Science Education and Research, Bhauri, Bhopal, Madhya Pradesh 462066, India
| | - Zeeshan Mushtaq
- Department of Biological Sciences, AB-3, Indian Institute of Science Education and Research, Bhauri, Bhopal, Madhya Pradesh 462066, India
| | - Manisha Kumari
- National Institute of Plant Genome Research (NIPGR), New Delhi 110067, India
| | - Praveen Kumar Verma
- National Institute of Plant Genome Research (NIPGR), New Delhi 110067, India
| | - Vimlesh Kumar
- Department of Biological Sciences, AB-3, Indian Institute of Science Education and Research, Bhauri, Bhopal, Madhya Pradesh 462066, India
| |
Collapse
|
39
|
Cortactin Is a Regulator of Activity-Dependent Synaptic Plasticity Controlled by Wingless. J Neurosci 2017; 37:2203-2215. [PMID: 28123080 DOI: 10.1523/jneurosci.1375-16.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 12/05/2016] [Accepted: 01/17/2017] [Indexed: 01/07/2023] Open
Abstract
Major signaling molecules initially characterized as key early developmental regulators are also essential for the plasticity of the nervous system. Previously, the Wingless (Wg)/Wnt pathway was shown to underlie the structural and electrophysiological changes during activity-dependent synaptic plasticity at the Drosophila neuromuscular junction. A challenge remains to understand how this signal mediates the cellular changes underlying this plasticity. Here, we focus on the actin regulator Cortactin, a major organizer of protrusion, membrane mobility, and invasiveness, and define its new role in synaptic plasticity. We show that Cortactin is present presynaptically and postsynaptically at the Drosophila NMJ and that it is a presynaptic regulator of rapid activity-dependent modifications in synaptic structure. Furthermore, animals lacking presynaptic Cortactin show a decrease in spontaneous release frequency, and presynaptic Cortactin is necessary for the rapid potentiation of spontaneous release frequency that takes place during activity-dependent plasticity. Most interestingly, Cortactin levels increase at stimulated synaptic terminals and this increase requires neuronal activity, de novo transcription and depends on Wg/Wnt expression. Because it is not simply the presence of Cortactin in the presynaptic terminal but its increase that is necessary for the full range of activity-dependent plasticity, we conclude that it probably plays a direct and important role in the regulation of this process.SIGNIFICANCE STATEMENT In the nervous system, changes in activity that lead to modifications in synaptic structure and function are referred to as synaptic plasticity and are thought to be the basis of learning and memory. The secreted Wingless/Wnt molecule is a potent regulator of synaptic plasticity in both vertebrates and invertebrates. Understanding the molecular mechanisms that underlie these plastic changes is a major gap in our knowledge. Here, we identify a presynaptic effector molecule of the Wingless/Wnt signal, Cortactin. We show that this molecule is a potent regulator of modifications in synaptic structure and is necessary for the electrophysiological changes taking place during synaptic plasticity.
Collapse
|
40
|
Xu Y, Quinn CC. SYD-1 Promotes Multiple Developmental Steps Leading to Neuronal Connectivity. Mol Neurobiol 2016; 53:6768-6773. [PMID: 26660112 PMCID: PMC5841450 DOI: 10.1007/s12035-015-9592-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 12/01/2015] [Indexed: 11/30/2022]
Abstract
The establishment of neuronal connectivity requires precise orchestration of multiple developmental steps, including axon specification, axon guidance, selection of synaptic target sites, and development of synaptic specializations. Although these are separate developmental steps, evidence indicates that some of the signaling molecules that regulate these steps are shared. In this review, we focus on SYD-1, a RhoGAP-like protein that has been implicated in each step of axonal development. We discuss interactions between SYD-1, UNC-40(DCC) and RhoGTPases and highlight both similarities and differences in how SYD-1 functions to regulate the different steps of axonal development. These observations reveal an example of how a signaling protein can be repurposed across sequential developmental steps.
Collapse
Affiliation(s)
- Yan Xu
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, 53211, USA
| | - Christopher C Quinn
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, 53211, USA.
| |
Collapse
|
41
|
Schulte C, Ripamonti M, Maffioli E, Cappelluti MA, Nonnis S, Puricelli L, Lamanna J, Piazzoni C, Podestà A, Lenardi C, Tedeschi G, Malgaroli A, Milani P. Scale Invariant Disordered Nanotopography Promotes Hippocampal Neuron Development and Maturation with Involvement of Mechanotransductive Pathways. Front Cell Neurosci 2016; 10:267. [PMID: 27917111 PMCID: PMC5114288 DOI: 10.3389/fncel.2016.00267] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 11/01/2016] [Indexed: 11/18/2022] Open
Abstract
The identification of biomaterials which promote neuronal maturation up to the generation of integrated neural circuits is fundamental for modern neuroscience. The development of neural circuits arises from complex maturative processes regulated by poorly understood signaling events, often guided by the extracellular matrix (ECM). Here we report that nanostructured zirconia surfaces, produced by supersonic cluster beam deposition of zirconia nanoparticles and characterized by ECM-like nanotopographical features, can direct the maturation of neural networks. Hippocampal neurons cultured on such cluster-assembled surfaces displayed enhanced differentiation paralleled by functional changes. The latter was demonstrated by single-cell electrophysiology showing earlier action potential generation and increased spontaneous postsynaptic currents compared to the neurons grown on the featureless unnaturally flat standard control surfaces. Label-free shotgun proteomics broadly confirmed the functional changes and suggests furthermore a vast impact of the neuron/nanotopography interaction on mechanotransductive machinery components, known to control physiological in vivo ECM-regulated axon guidance and synaptic plasticity. Our results indicate a potential of cluster-assembled zirconia nanotopography exploitable for the creation of efficient neural tissue interfaces and cell culture devices promoting neurogenic events, but also for unveiling mechanotransductive aspects of neuronal development and maturation.
Collapse
Affiliation(s)
- Carsten Schulte
- Dipartimento di Fisica, Centro Interdisciplinare Materiali e Interfacce Nanostrutturate, Università degli Studi di MilanoMilan, Italy; Fondazione FilareteMilan, Italy
| | - Maddalena Ripamonti
- Neurobiology of Learning Unit, Division of Neuroscience, Scientific Institute San Raffaele, Università Vita-Salute San Raffaele Milan, Italy
| | - Elisa Maffioli
- Fondazione FilareteMilan, Italy; Dipartimento di Medicina Veterinaria, Università degli Studi di MilanoMilan, Italy
| | - Martino A Cappelluti
- Fondazione FilareteMilan, Italy; SEMM - European School of Molecular MedicineMilan, Italy
| | - Simona Nonnis
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano Milan, Italy
| | - Luca Puricelli
- Dipartimento di Fisica, Centro Interdisciplinare Materiali e Interfacce Nanostrutturate, Università degli Studi di Milano Milan, Italy
| | - Jacopo Lamanna
- Neurobiology of Learning Unit, Division of Neuroscience, Scientific Institute San Raffaele, Università Vita-Salute San Raffaele Milan, Italy
| | - Claudio Piazzoni
- Dipartimento di Fisica, Centro Interdisciplinare Materiali e Interfacce Nanostrutturate, Università degli Studi di Milano Milan, Italy
| | - Alessandro Podestà
- Dipartimento di Fisica, Centro Interdisciplinare Materiali e Interfacce Nanostrutturate, Università degli Studi di Milano Milan, Italy
| | - Cristina Lenardi
- Dipartimento di Fisica, Centro Interdisciplinare Materiali e Interfacce Nanostrutturate, Università degli Studi di Milano Milan, Italy
| | - Gabriella Tedeschi
- Fondazione FilareteMilan, Italy; Dipartimento di Medicina Veterinaria, Università degli Studi di MilanoMilan, Italy
| | - Antonio Malgaroli
- Neurobiology of Learning Unit, Division of Neuroscience, Scientific Institute San Raffaele, Università Vita-Salute San Raffaele Milan, Italy
| | - Paolo Milani
- Dipartimento di Fisica, Centro Interdisciplinare Materiali e Interfacce Nanostrutturate, Università degli Studi di Milano Milan, Italy
| |
Collapse
|
42
|
Mechanisms controlling assembly and plasticity of presynaptic active zone scaffolds. Curr Opin Neurobiol 2016; 39:69-76. [DOI: 10.1016/j.conb.2016.04.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 04/08/2016] [Accepted: 04/15/2016] [Indexed: 11/18/2022]
|
43
|
Stavoe AKH, Kargbo-Hill SE, Hall DH, Colón-Ramos DA. KIF1A/UNC-104 Transports ATG-9 to Regulate Neurodevelopment and Autophagy at Synapses. Dev Cell 2016; 38:171-85. [PMID: 27396362 PMCID: PMC4961624 DOI: 10.1016/j.devcel.2016.06.012] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 05/09/2016] [Accepted: 06/09/2016] [Indexed: 01/12/2023]
Abstract
Autophagy is a cellular degradation process important for neuronal development and survival. Neurons are highly polarized cells in which autophagosome biogenesis is spatially compartmentalized. The mechanisms and physiological importance of this spatial compartmentalization of autophagy in the neuronal development of living animals are not well understood. Here we determine that, in Caenorhabditis elegans neurons, autophagosomes form near synapses and are required for neurodevelopment. We first determine, through unbiased genetic screens and systematic genetic analyses, that autophagy is required cell autonomously for presynaptic assembly and for axon outgrowth dynamics in specific neurons. We observe autophagosome biogenesis in the axon near synapses, and this localization depends on the synaptic vesicle kinesin, KIF1A/UNC-104. KIF1A/UNC-104 coordinates localized autophagosome formation by regulating the transport of the integral membrane autophagy protein, ATG-9. Our findings indicate that autophagy is spatially regulated in neurons through the transport of ATG-9 by KIF1A/UNC-104 to regulate neurodevelopment.
Collapse
Affiliation(s)
- Andrea K H Stavoe
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Departments of Cell Biology and Neuroscience, Yale University School of Medicine, 295 Congress Avenue, BCMM 436B, New Haven, CT 06536-0812, USA
| | - Sarah E Kargbo-Hill
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Departments of Cell Biology and Neuroscience, Yale University School of Medicine, 295 Congress Avenue, BCMM 436B, New Haven, CT 06536-0812, USA
| | - David H Hall
- Center for C. elegans Anatomy, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Daniel A Colón-Ramos
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Departments of Cell Biology and Neuroscience, Yale University School of Medicine, 295 Congress Avenue, BCMM 436B, New Haven, CT 06536-0812, USA; Instituto de Neurobiología, Recinto de Ciencias Médicas, Universidad de Puerto Rico, 201 Boulevard del Valle, San Juan 00901, Puerto Rico.
| |
Collapse
|
44
|
Schröter S, Beckmann S, Schmitt HD. ER arrival sites for COPI vesicles localize to hotspots of membrane trafficking. EMBO J 2016; 35:1935-55. [PMID: 27440402 DOI: 10.15252/embj.201592873] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 06/21/2016] [Indexed: 11/09/2022] Open
Abstract
COPI-coated vesicles mediate retrograde membrane traffic from the cis-Golgi to the endoplasmic reticulum (ER) in all eukaryotic cells. However, it is still unknown whether COPI vesicles fuse everywhere or at specific sites with the ER membrane. Taking advantage of the circumstance that the vesicles still carry their coat when they arrive at the ER, we have visualized active ER arrival sites (ERAS) by monitoring contact between COPI coat components and the ER-resident Dsl tethering complex using bimolecular fluorescence complementation (BiFC). ERAS form punctate structures near Golgi compartments, clearly distinct from ER exit sites. Furthermore, ERAS are highly polarized in an actin and myosin V-dependent manner and are localized near hotspots of plasma membrane expansion. Genetic experiments suggest that the COPI•Dsl BiFC complexes recapitulate the physiological interaction between COPI and the Dsl complex and that COPI vesicles are mistargeted in dsl1 mutants. We conclude that the Dsl complex functions in confining COPI vesicle fusion sites.
Collapse
Affiliation(s)
- Saskia Schröter
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Sabrina Beckmann
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Hans Dieter Schmitt
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| |
Collapse
|
45
|
Pinto MJ, Pedro JR, Costa RO, Almeida RD. Visualizing K48 Ubiquitination during Presynaptic Formation By Ubiquitination-Induced Fluorescence Complementation (UiFC). Front Mol Neurosci 2016; 9:43. [PMID: 27375430 PMCID: PMC4901079 DOI: 10.3389/fnmol.2016.00043] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/24/2016] [Indexed: 11/18/2022] Open
Abstract
In recent years, signaling through ubiquitin has been shown to be of great importance for normal brain development. Indeed, fluctuations in ubiquitin levels and spontaneous mutations in (de)ubiquitination enzymes greatly perturb synapse formation and neuronal transmission. In the brain, expression of lysine (K) 48-linked ubiquitin chains is higher at a developmental stage coincident with synaptogenesis. Nevertheless, no studies have so far delved into the involvement of this type of polyubiquitin chains in synapse formation. We have recently proposed a role for polyubiquitinated conjugates as triggering signals for presynaptic assembly. Herein, we aimed at characterizing the axonal distribution of K48 polyubiquitin and its dynamics throughout the course of presynaptic formation. To accomplish so, we used an ubiquitination-induced fluorescence complementation (UiFC) strategy for the visualization of K48 polyubiquitin in live hippocampal neurons. We first validated its use in neurons by analyzing changing levels of polyubiquitin. UiFC signal is diffusely distributed with distinct aggregates in somas, dendrites and axons, which perfectly colocalize with staining for a K48-specific antibody. Axonal UiFC aggregates are relatively stable and new aggregates are formed as an axon grows. Approximately 65% of UiFC aggregates colocalize with synaptic vesicle clusters and they preferentially appear in the axonal domains of axo-somatodendritic synapses when compared to isolated axons. We then evaluated axonal accumulation of K48 ubiquitinated signals in bead-induced synapses. We observed rapid accumulation of UiFC signal and endogenous K48 ubiquitin at the sites of newly formed presynapses. Lastly, we show by means of a microfluidic platform, for the isolation of axons, that presynaptic clustering on beads is dependent on E1-mediated ubiquitination at the axonal level. Altogether, these results indicate that enrichment of K48 polyubiquitin at the site of nascent presynaptic terminals is an important axon-intrinsic event for presynaptic differentiation.
Collapse
Affiliation(s)
- Maria J Pinto
- Center for Neuroscience and Cell Biology (CNC), University of CoimbraCoimbra, Portugal; PhD Programme in Experimental Biology and Biomedicine (PDBEB), Center for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal
| | - Joana R Pedro
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra Coimbra, Portugal
| | - Rui O Costa
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra Coimbra, Portugal
| | - Ramiro D Almeida
- Center for Neuroscience and Cell Biology (CNC), University of CoimbraCoimbra, Portugal; School of Allied Health Technologies, Polytechnic Institute of Porto (ESTSP-IPP)Vila Nova de Gaia, Portugal; Institute for Interdisciplinary Research, University of CoimbraCoimbra, Portugal
| |
Collapse
|
46
|
Abstract
Elimination of the excess synaptic contacts established in the early stages of neuronal development is required to refine the function of neuronal circuits. Here we investigate whether secreted protein acidic and rich in cysteine (SPARC), a molecule produced by glial cells, is involved in synapse removal. SPARC production peaks when innervation of the rat superior cervical ganglion and the tail of Xenopus tropicalis tadpoles are remodeled. The formation of new cholinergic synapses in autaptic single-cell microcultures is inhibited by SPARC. The effect resides in the C-terminal domain, which is also responsible for triggering a concentration- and time-dependent disassembly of stable cholinergic synapses. The loss of synaptic contacts is associated with the formation of retracted axon terminals containing multivesicular bodies and secondary lysosomes. The biological relevance of in vitro results was supported by injecting the tail of Xenopus tropicalis tadpoles with peptide 4.2, a 20-aa sequence derived from SPARC that mimics full-length protein effects. Swimming was severely impaired at ∼5 h after peptide application, caused by the massive elimination of neuromuscular junctions and pruning of axonal branches. Effects revert by 6 d after injection, as motor innervation reforms. In conclusion, SPARC triggers a cell-autonomous program of synapse elimination in cholinergic neurons that likely occurs when protein production peaks during normal development.
Collapse
|
47
|
Kevenaar JT, Hoogenraad CC. The axonal cytoskeleton: from organization to function. Front Mol Neurosci 2015; 8:44. [PMID: 26321907 PMCID: PMC4536388 DOI: 10.3389/fnmol.2015.00044] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 07/31/2015] [Indexed: 01/20/2023] Open
Abstract
The axon is the single long fiber that extends from the neuron and transmits electrical signals away from the cell body. The neuronal cytoskeleton, composed of microtubules (MTs), actin filaments and neurofilaments, is not only required for axon formation and axonal transport but also provides the structural basis for several specialized axonal structures, such as the axon initial segment (AIS) and presynaptic boutons. Emerging evidence suggest that the unique cytoskeleton organization in the axon is essential for its structure and integrity. In addition, the increasing number of neurodevelopmental and neurodegenerative diseases linked to defect in actin- and microtubule-dependent processes emphasizes the importance of a properly regulated cytoskeleton for normal axonal functioning. Here, we provide an overview of the current understanding of actin and microtubule organization within the axon and discuss models for the functional role of the cytoskeleton at specialized axonal structures.
Collapse
Affiliation(s)
- Josta T. Kevenaar
- Cell Biology, Faculty of Science, Utrecht UniversityUtrecht, Netherlands
| | | |
Collapse
|
48
|
Comparative Proteomic Analysis of Carbonylated Proteins from the Striatum and Cortex of Pesticide-Treated Mice. PARKINSONS DISEASE 2015; 2015:812532. [PMID: 26345149 PMCID: PMC4546751 DOI: 10.1155/2015/812532] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 05/27/2015] [Accepted: 05/30/2015] [Indexed: 01/28/2023]
Abstract
Epidemiological studies indicate exposures to the herbicide paraquat (PQ) and fungicide maneb (MB) are associated with increased risk of Parkinson's disease (PD). Oxidative stress appears to be a premier mechanism that underlies damage to the nigrostriatal dopamine system in PD and pesticide exposure. Enhanced oxidative stress leads to lipid peroxidation and production of reactive aldehydes; therefore, we conducted proteomic analyses to identify carbonylated proteins in the striatum and cortex of pesticide-treated mice in order to elucidate possible mechanisms of toxicity. Male C57BL/6J mice were treated biweekly for 6 weeks with saline, PQ (10 mg/kg), MB (30 mg/kg), or the combination of PQ and MB (PQMB). Treatments resulted in significant behavioral alterations in all treated mice and depleted striatal dopamine in PQMB mice. Distinct differences in 4-hydroxynonenal-modified proteins were observed in the striatum and cortex. Proteomic analyses identified carbonylated proteins and peptides from the cortex and striatum, and pathway analyses revealed significant enrichment in a variety of KEGG pathways. Further analysis showed enrichment in proteins of the actin cytoskeleton in treated samples, but not in saline controls. These data indicate that treatment-related effects on cytoskeletal proteins could alter proper synaptic function, thereby resulting in impaired neuronal function and even neurodegeneration.
Collapse
|
49
|
Ganguly A, Tang Y, Wang L, Ladt K, Loi J, Dargent B, Leterrier C, Roy S. A dynamic formin-dependent deep F-actin network in axons. J Cell Biol 2015. [PMID: 26216902 PMCID: PMC4523607 DOI: 10.1083/jcb.201506110] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Although actin at neuronal growth cones is well-studied, much less is known about actin organization and dynamics along axon shafts and presynaptic boutons. Using probes that selectively label filamentous-actin (F-actin), we found focal "actin hotspots" along axons-spaced ∼3-4 µm apart-where actin undergoes continuous assembly/disassembly. These foci are a nidus for vigorous actin polymerization, generating long filaments spurting bidirectionally along axons-a phenomenon we call "actin trails." Super-resolution microscopy reveals intra-axonal deep actin filaments in addition to the subplasmalemmal "actin rings" described recently. F-actin hotspots colocalize with stationary axonal endosomes, and blocking vesicle transport diminishes the actin trails, suggesting mechanistic links between vesicles and F-actin kinetics. Actin trails are formin-but not Arp2/3-dependent and help enrich actin at presynaptic boutons. Finally, formin inhibition dramatically disrupts synaptic recycling. Collectively, available data suggest a two-tier F-actin organization in axons, with stable "actin rings" providing mechanical support to the plasma membrane and dynamic "actin trails" generating a flexible cytoskeletal network with putative physiological roles.
Collapse
Affiliation(s)
- Archan Ganguly
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093
| | - Yong Tang
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093
| | - Lina Wang
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093
| | - Kelsey Ladt
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093 Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093
| | - Jonathan Loi
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093
| | - Bénédicte Dargent
- Aix Marseille Université, Centre National de la Recherche Scientifique, Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille (CRN2M) UMR7286, 13344 Marseille, France
| | - Christophe Leterrier
- Aix Marseille Université, Centre National de la Recherche Scientifique, Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille (CRN2M) UMR7286, 13344 Marseille, France
| | - Subhojit Roy
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093 Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
50
|
Matsumoto N, Hoshiko M, Sugo N, Fukazawa Y, Yamamoto N. Synapse-dependent and independent mechanisms of thalamocortical axon branching are regulated by neuronal activity. Dev Neurobiol 2015; 76:323-36. [PMID: 26061995 DOI: 10.1002/dneu.22317] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 06/06/2015] [Indexed: 12/23/2022]
Abstract
Axon branching and synapse formation are critical processes for establishing precise circuit connectivity. These processes are tightly regulated by neural activity, but the relationship between them remains largely unclear. We use organotypic coculture preparations to examine the role of synapse formation in the activity-dependent axon branching of thalamocortical (TC) projections. To visualize TC axons and their presynaptic sites, two plasmids encoding DsRed and EGFP-tagged synaptophysin (SYP-EGFP) were cotransfected into a small number of thalamic neurons. Time-lapse imaging of individual TC axons showed that most branches emerged from SYP-EGFP puncta, indicating that synapse formation precedes emergences of axonal branches. We also investigated the effects of neuronal activity on axon branching and synapse formation by manipulating spontaneous firing activity of thalamic cells. An inward rectifying potassium channel, Kir2.1, and a bacterial voltage-gated sodium channel, NaChBac, were used to suppress and promote firing activity, respectively. We found suppressing neural activity reduced both axon branching and synapse formation. In contrast, increasing neural activity promoted only axonal branch formation. Time-lapse imaging of NaChBac-expressing cells further revealed that new branches frequently appeared from the locations other than SYP-EGFP puncta, indicating that enhancing activity promotes axonal branch formation due to an increase of branch emergence at nonsynaptic sites. These results suggest that presynaptic locations are hotspots for branch emergence, and that frequent firing activity can shift branch emergence to a synapse-independent process.
Collapse
Affiliation(s)
- Naoyuki Matsumoto
- Laboratory of Cellular and Molecular Neurobiology Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Maki Hoshiko
- Laboratory of Cellular and Molecular Neurobiology Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Noriyuki Sugo
- Laboratory of Cellular and Molecular Neurobiology Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yugo Fukazawa
- Division of Brain Structure and Function, Faculty of Medical Science, University of Fukui, Eiheiji, Yoshida, 910-1193, Japan
| | - Nobuhiko Yamamoto
- Laboratory of Cellular and Molecular Neurobiology Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
| |
Collapse
|