1
|
Bruentgens F, Moreno Velasquez L, Stumpf A, Parthier D, Breustedt J, Benfenati F, Milovanovic D, Schmitz D, Orlando M. The Lack of Synapsin Alters Presynaptic Plasticity at Hippocampal Mossy Fibers in Male Mice. eNeuro 2024; 11:ENEURO.0330-23.2024. [PMID: 38866497 PMCID: PMC11223178 DOI: 10.1523/eneuro.0330-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/14/2024] Open
Abstract
Synapsins are highly abundant presynaptic proteins that play a crucial role in neurotransmission and plasticity via the clustering of synaptic vesicles. The synapsin III isoform is usually downregulated after development, but in hippocampal mossy fiber boutons, it persists in adulthood. Mossy fiber boutons express presynaptic forms of short- and long-term plasticity, which are thought to underlie different forms of learning. Previous research on synapsins at this synapse focused on synapsin isoforms I and II. Thus, a complete picture regarding the role of synapsins in mossy fiber plasticity is still missing. Here, we investigated presynaptic plasticity at hippocampal mossy fiber boutons by combining electrophysiological field recordings and transmission electron microscopy in a mouse model lacking all synapsin isoforms. We found decreased short-term plasticity, i.e., decreased facilitation and post-tetanic potentiation, but increased long-term potentiation in male synapsin triple knock-out (KO) mice. At the ultrastructural level, we observed more dispersed vesicles and a higher density of active zones in mossy fiber boutons from KO animals. Our results indicate that all synapsin isoforms are required for fine regulation of short- and long-term presynaptic plasticity at the mossy fiber synapse.
Collapse
Affiliation(s)
- Felicitas Bruentgens
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Laura Moreno Velasquez
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Alexander Stumpf
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Daniel Parthier
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
| | - Jörg Breustedt
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genoa 16163, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa 16132, Italy
| | - Dragomir Milovanovic
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin 10117, Germany
- Einstein Center for Neurosciences, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin 10117, Germany
| | - Dietmar Schmitz
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin 10117, Germany
- Einstein Center for Neurosciences, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin 10117, Germany
- Bernstein Center for Computational Neuroscience, Humboldt-Universität zu Berlin, Berlin 10115, Germany
| | - Marta Orlando
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| |
Collapse
|
2
|
Takahashi H, Hisata K, Iguchi R, Kikuchi S, Ogasawara M, Satoh N. scRNA-seq analysis of cells comprising the amphioxus notochord. Dev Biol 2024; 508:24-37. [PMID: 38224933 DOI: 10.1016/j.ydbio.2024.01.003] [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: 09/25/2023] [Revised: 01/04/2024] [Accepted: 01/06/2024] [Indexed: 01/17/2024]
Abstract
Cephalochordates occupy a key phylogenetic position for deciphering the origin and evolution of chordates, since they diverged earlier than urochordates and vertebrates. The notochord is the most prominent feature of chordates. The amphioxus notochord features coin-shaped cells bearing myofibrils. Notochord-derived hedgehog signaling contributes to patterning of the dorsal nerve cord, as in vertebrates. However, properties of constituent notochord cells remain unknown at the single-cell level. We examined these properties using Iso-seq analysis, single-cell RNA-seq analysis, and in situ hybridization (ISH). Gene expression profiles broadly categorize notochordal cells into myofibrillar cells and non-myofibrillar cells. Myofibrillar cells occupy most of the central portion of the notochord, and some cells extend the notochordal horn to both sides of the ventral nerve cord. Some notochord myofibrillar genes are not expressed in myotomes, suggesting an occurrence of myofibrillar genes that are preferentially expressed in notochord. On the other hand, non-myofibrillar cells contain dorsal, lateral, and ventral Müller cells, and all three express both hedgehog and Brachyury. This was confirmed by ISH, although expression of hedgehog in ventral Müller cells was minimal. In addition, dorsal Müller cells express neural transmission-related genes, suggesting an interaction with nerve cord. Lateral Müller cells express hedgehog and other signaling-related genes, suggesting an interaction with myotomes positioned lateral to the notochord. Ventral Müller cells also expressed genes for FGF- and EGF-related signaling, which may be associated with development of endoderm, ventral to the notochord. Lateral Müller cells were intermediate between dorsal/ventral Müller cells. Since vertebrate notochord contributes to patterning and differentiation of ectoderm (nerve cord), mesoderm (somite), and endoderm, this investigation provides evidence that an ancestral or original form of vertebrate notochord is present in extant cephalochordates.
Collapse
Affiliation(s)
- Hiroki Takahashi
- Interdisciplinary Research Unit, National Institute for Basic Biology, Okazaki, Aichi, 444-8585, Japan.
| | - Kanako Hisata
- Marine Genomics Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa, 904-0495, Japan
| | - Rin Iguchi
- Department of Biology, Graduate School of Science, Chiba University, Chiba, 262-8522, Japan
| | - Sakura Kikuchi
- Marine Genomics Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa, 904-0495, Japan
| | - Michio Ogasawara
- Department of Biology, Graduate School of Science, Chiba University, Chiba, 262-8522, Japan.
| | - Noriyuki Satoh
- Marine Genomics Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa, 904-0495, Japan.
| |
Collapse
|
3
|
Baris RO, Sahin N, Bilgic AD, Ozdemir C, Edgunlu TG. Molecular and in silico analyses of SYN III gene variants in autism spectrum disorder. Ir J Med Sci 2023; 192:2887-2895. [PMID: 37166614 DOI: 10.1007/s11845-023-03402-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/04/2023] [Indexed: 05/12/2023]
Abstract
BACKGROUND Defects in neurotransmission and synaptogenesis are noteworthy in the pathogenesis of ASD. Synapsin III (SYN III) is defined as a synaptic vesicle protein that plays an important role in synaptogenesis and regulation of neurotransmitter release and neurite outgrowth. Therefore, SYN III may associate with many neurodevelopmental diseases, including ASD. AIM The aim of this study was to investigate whether the SYN III gene -631 C > G (rs133946) and -196 G > A (rs133945) polymorphisms are associated with susceptibility to ASD. METHODS SYN III variants and the risk of ASD were investigated in 26 healthy children and 24 ASD children. SYN III gene variants were genotyped by PCR-RFLP methods. The differences in genotype and allele frequencies between the ASD and control groups were calculated using the chi-square (χ2). We analysed the SYN III gene using web-based tools. RESULTS Our results suggest that the presence of the AA genotype of the SYN III -196 G > A (rs133945) polymorphism affects the characteristics and development of ASD in children (p = 0.012). SYN III -631 C > G (rs133946) polymorphism was not associated with ASD (p = 0.524). We have shown the prediction of gene-gene interaction that SYN III is co-expressed with 17 genes, physical interaction with 3 genes, and co-localization with 12 genes. The importance of different genes (SYN I, II, III, GABRD, NOS1AP, GNAO1) for ASD pathogenesis was revealed by GO analysis. CONCLUSION Considering the role of SYN III and related genes, especially in the synaptic vesicle pathway and neurotransmission, its effect on ASD can be further investigated.
Collapse
Affiliation(s)
- Remzi Oguz Baris
- Faculty of Medicine, Mugla Sitki Kocman University, Mugla, Turkey
| | - Nilfer Sahin
- Department of Child and Adolescent Mental Health Diseases School of Medicine, Muğla Sıtkı Koçman University, Mugla, Turkey
| | - Ayşegül Demirtas Bilgic
- Department of Medical Biology, Health Sciences Institution, Muğla Sıtkı Koçman University, Mugla, Turkey
| | - Cilem Ozdemir
- Department of Medical Biology, Health Sciences Institution, Muğla Sıtkı Koçman University, Mugla, Turkey.
| | - Tuba Gokdogan Edgunlu
- Department of Medical Biology, School of Medicine, Muğla Sıtkı Koçman University, Mugla, 48000, Turkey
| |
Collapse
|
4
|
Longhena F, Faustini G, Brembati V, Pizzi M, Benfenati F, Bellucci A. An updated reappraisal of synapsins: structure, function and role in neurological and psychiatric disorders. Neurosci Biobehav Rev 2021; 130:33-60. [PMID: 34407457 DOI: 10.1016/j.neubiorev.2021.08.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/29/2021] [Accepted: 08/09/2021] [Indexed: 01/02/2023]
Abstract
Synapsins (Syns) are phosphoproteins strongly involved in neuronal development and neurotransmitter release. Three distinct genes SYN1, SYN2 and SYN3, with elevated evolutionary conservation, have been described to encode for Synapsin I, Synapsin II and Synapsin III, respectively. Syns display a series of common features, but also exhibit distinctive localization, expression pattern, post-translational modifications (PTM). These characteristics enable their interaction with other synaptic proteins, membranes and cytoskeletal components, which is essential for the proper execution of their multiple functions in neuronal cells. These include the control of synapse formation and growth, neuron maturation and renewal, as well as synaptic vesicle mobilization, docking, fusion, recycling. Perturbations in the balanced expression of Syns, alterations of their PTM, mutations and polymorphisms of their encoding genes induce severe dysregulations in brain networks functions leading to the onset of psychiatric or neurological disorders. This review presents what we have learned since the discovery of Syn I in 1977, providing the state of the art on Syns structure, function, physiology and involvement in central nervous system disorders.
Collapse
Affiliation(s)
- Francesca Longhena
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy.
| | - Gaia Faustini
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy.
| | - Viviana Brembati
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy.
| | - Marina Pizzi
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy.
| | - Fabio Benfenati
- Italian Institute of Technology, Via Morego 30, Genova, Italy; IRCSS Policlinico San Martino Hospital, Largo Rosanna Benzi 10, 16132, Genova, Italy.
| | - Arianna Bellucci
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy; Laboratory for Preventive and Personalized Medicine, Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy.
| |
Collapse
|
5
|
Wu CC, Brugeaud A, Seist R, Lin HC, Yeh WH, Petrillo M, Coppola G, Edge ASB, Stankovic KM. Altered expression of genes regulating inflammation and synaptogenesis during regrowth of afferent neurons to cochlear hair cells. PLoS One 2020; 15:e0238578. [PMID: 33001981 PMCID: PMC7529247 DOI: 10.1371/journal.pone.0238578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 08/19/2020] [Indexed: 12/20/2022] Open
Abstract
The spiral ganglion neurons constitute the primary connection between auditory hair cells and the brain. The spiral ganglion afferent fibers and their synapse with hair cells do not regenerate to any significant degree in adult mammalian ears after damage. We have investigated gene expression changes after kainate-induced disruption of the synapses in a neonatal cochlear explant model in which peripheral fibers and the afferent synapse do regenerate. We compared gene expression early after damage, during regeneration of the fibers and synapses, and after completion of in vitro regeneration. These analyses revealed a total of 2.5% differentially regulated transcripts (588 out of 24,000) based on a threshold of p<0.005. Inflammatory response genes as well as genes involved in regeneration of neural circuits were upregulated in the spiral ganglion neurons and organ of Corti, where the hair cells reside. Prominent genes upregulated at several time points included genes with roles in neurogenesis (Elavl4 and Sox21), neural outgrowth (Ntrk3 and Ppp1r1c), axonal guidance (Rgmb and Sema7a), synaptogenesis (Nlgn2 and Psd2), and synaptic vesicular function (Syt8 and Syn1). Immunohistochemical and in situ hybridization analysis of genes that had not previously been described in the cochlea confirmed their cochlear expression. The time course of expression of these genes suggests that kainate treatment resulted in a two-phase response in spiral ganglion neurons: an acute response consistent with inflammation, followed by an upregulation of neural regeneration genes. Identification of the genes activated during regeneration of these fibers suggests candidates that could be targeted to enhance regeneration in adult ears.
Collapse
Affiliation(s)
- Chen-Chi Wu
- Eaton Peabody Laboratories and Department of Otolaryngology—Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Department of Otolaryngology—Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Aurore Brugeaud
- Eaton Peabody Laboratories and Department of Otolaryngology—Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Department of Otolaryngology—Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Richard Seist
- Eaton Peabody Laboratories and Department of Otolaryngology—Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Department of Otolaryngology—Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Otorhinolaryngology-Head and Neck Surgery, Paracelsus Medical University, Salzburg, Austria
| | - Hsiao-Chun Lin
- Eaton Peabody Laboratories and Department of Otolaryngology—Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Department of Otolaryngology—Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Wei-Hsi Yeh
- Eaton Peabody Laboratories and Department of Otolaryngology—Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Department of Otolaryngology—Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Marco Petrillo
- Eaton Peabody Laboratories and Department of Otolaryngology—Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Department of Otolaryngology—Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Giovanni Coppola
- Program in Neurogenetics, Department of Neurology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Albert S. B. Edge
- Eaton Peabody Laboratories and Department of Otolaryngology—Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Department of Otolaryngology—Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Konstantina M. Stankovic
- Eaton Peabody Laboratories and Department of Otolaryngology—Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Department of Otolaryngology—Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, Massachusetts, United States of America
- Harvard Program in Therapeutic Science, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
6
|
Huebschman JL, Corona KS, Guo Y, Smith LN. The Fragile X Mental Retardation Protein Regulates Striatal Medium Spiny Neuron Synapse Density and Dendritic Spine Morphology. Front Mol Neurosci 2020; 13:161. [PMID: 33013316 PMCID: PMC7511717 DOI: 10.3389/fnmol.2020.00161] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/05/2020] [Indexed: 01/23/2023] Open
Abstract
The fragile X mental retardation protein (FMRP), an RNA-binding protein that mediates the transport, stability, and translation of hundreds of brain RNAs, is critically involved in regulating synaptic function. Loss of FMRP, as in fragile X syndrome (FXS), is a leading monogenic cause of autism and results in altered structural and functional synaptic plasticity, widely described in the hippocampus and cortex. Though FXS is associated with hyperactivity, impaired social interaction, and the development of repetitive or stereotyped behaviors, all of which are influenced by striatal activity, few studies have investigated the function of FMRP here. Utilizing a cortical-striatal co-culture model, we find that striatal medium spiny neurons (MSNs) lacking FMRP fail to make normal increases in PSD95 expression over a short time period and have significant deficits in dendritic spine density and colocalized synaptic puncta at the later measured time point compared to wildtype (WT) MSNs. Acute expression of wtFMRP plasmid in Fmr1 KO co-cultures results in contrasting outcomes for these measures on MSNs at the more mature time point, reducing spine density across multiple spine types but making no significant changes in colocalized puncta. FMRP’s KH2 and RGG RNA-binding domains are required for normal elimination of PSD95, and interruption of these domains slightly favors elimination of immature spine types. Further, KH2 is required for normal levels of colocalized puncta. Our data are largely consistent with a basal role for FMRP and its RNA-binding domains in striatal synapse stabilization on developing MSNs, and in light of previous findings, suggest distinct regional and/or cell type-specific roles for FMRP in regulating synapse structure.
Collapse
Affiliation(s)
- Jessica L Huebschman
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, United States.,Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, United States
| | - Kitzia S Corona
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Yuhong Guo
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Laura N Smith
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, United States.,Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, United States
| |
Collapse
|
7
|
Ma M, Xiong W, Hu F, Deng MF, Huang X, Chen JG, Man HY, Lu Y, Liu D, Zhu LQ. A novel pathway regulates social hierarchy via lncRNA AtLAS and postsynaptic synapsin IIb. Cell Res 2020; 30:105-118. [PMID: 31959917 DOI: 10.1038/s41422-020-0273-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 12/30/2019] [Indexed: 12/16/2022] Open
Abstract
Dominance hierarchy is a fundamental phenomenon in grouped animals and human beings, however, the underlying regulatory mechanisms remain elusive. Here, we report that an antisense long non-coding RNA (lncRNA) of synapsin II, named as AtLAS, plays a crucial role in the regulation of social hierarchy. AtLAS is decreased in the prefrontal cortical excitatory pyramidal neurons of dominant mice; consistently, silencing or overexpression of AtLAS increases or decreases the social rank, respectively. Mechanistically, we show that AtLAS regulates alternative polyadenylation of synapsin II gene and increases synapsin 2b (syn2b) expression. Syn2b reduces AMPA receptor (AMPAR)-mediated excitatory synaptic transmission through a direct binding with AMPAR at the postsynaptic site via its unique C-terminal sequence. Moreover, a peptide disrupting the binding of syn2b with AMPARs enhances the synaptic strength and social ranks. These findings reveal a novel role for lncRNA AtLAS and its target syn2b in the regulation of social behaviors by controlling postsynaptic AMPAR trafficking.
Collapse
Affiliation(s)
- Mei Ma
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Wan Xiong
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Fan Hu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Man-Fei Deng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Xian Huang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jian-Guo Chen
- The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA, 02215, USA
| | - Youming Lu
- The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Dan Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China. .,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Ling-Qiang Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China. .,The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
8
|
Wesseling JF, Phan S, Bushong EA, Siksou L, Marty S, Pérez-Otaño I, Ellisman M. Sparse force-bearing bridges between neighboring synaptic vesicles. Brain Struct Funct 2019; 224:3263-3276. [PMID: 31667576 PMCID: PMC6875159 DOI: 10.1007/s00429-019-01966-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: 06/10/2019] [Accepted: 10/05/2019] [Indexed: 12/28/2022]
Abstract
Most vesicles in the interior of synaptic terminals are clustered in clouds close to active zone regions of the plasma membrane where exocytosis occurs. Electron-dense structures, termed bridges, have been reported between a small minority of pairs of neighboring vesicles within the clouds. Synapsin proteins have been implicated previously, but the existence of the bridges as stable structures in vivo has been questioned. Here we use electron tomography to show that the bridges are present but less frequent in synapsin knockouts compared to wildtype. An analysis of distances between neighbors in wildtype tomograms indicated that the bridges are strong enough to resist centrifugal forces likely induced by fixation with aldehydes. The results confirm that the bridges are stable structures and that synapsin proteins are involved in formation or stabilization.
Collapse
Affiliation(s)
- John F Wesseling
- Instituto de Neurociencias, CSIC-UMH, San Juan de Alicante, Spain. .,Departmento de Neurociencias (CIMA), Universidad de Navarra, Pamplona, Spain.
| | - Sébastien Phan
- National Center for Microscopy and Imaging Research, University of California, San Diego, CA, USA
| | - Eric A Bushong
- National Center for Microscopy and Imaging Research, University of California, San Diego, CA, USA
| | - Léa Siksou
- Institut de Biologie de l'ENS (IBENS), École Normale Supérieure, INSERM, CNRS, PSL Research University, Paris, France.,Global Research and Development, Teva Pharmaceutical Industries Ltd, Netanya, Israel
| | - Serge Marty
- Institut de Biologie de l'ENS (IBENS), École Normale Supérieure, INSERM, CNRS, PSL Research University, Paris, France.,Institut du Cerveau et de la Moelle épinière, INSERM U1127, CNRS UMR7225, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | | | - Mark Ellisman
- National Center for Microscopy and Imaging Research and Department of Neuroscience, University of California, San Diego, CA, USA
| |
Collapse
|
9
|
Affiliation(s)
- Laura E Perlini
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova, Italy
| | - Fabio Benfenati
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
| | - Laura Cancedda
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova, Italy.,Dulbecco Telethon Institute, Genova, Italy
| |
Collapse
|
10
|
Cheng Q, Song SH, Augustine GJ. Molecular Mechanisms of Short-Term Plasticity: Role of Synapsin Phosphorylation in Augmentation and Potentiation of Spontaneous Glutamate Release. Front Synaptic Neurosci 2018; 10:33. [PMID: 30425632 PMCID: PMC6218601 DOI: 10.3389/fnsyn.2018.00033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/20/2018] [Indexed: 12/24/2022] Open
Abstract
We used genetic and pharmacological approaches to identify the signaling pathways involved in augmentation and potentiation, two forms of activity dependent, short-term synaptic plasticity that enhance neurotransmitter release. Trains of presynaptic action potentials produced a robust increase in the frequency of miniature excitatory postsynaptic currents (mEPSCs). Following the end of the stimulus, mEPSC frequency followed a bi-exponential decay back to basal levels. The time constants of decay identified these two exponential components as the decay of augmentation and potentiation, respectively. Augmentation increased mEPSC frequency by 9.3-fold, while potentiation increased mEPSC frequency by 2.4-fold. In synapsin triple-knockout (TKO) neurons, augmentation was reduced by 83% and potentiation was reduced by 74%, suggesting that synapsins are key signaling elements in both forms of plasticity. To examine the synapsin isoforms involved, we expressed individual synapsin isoforms in TKO neurons. While synapsin IIIa rescued both augmentation and potentiation, none of the other synapsin isoforms produced statistically significant amounts of rescue. To determine the involvement of protein kinases in these two forms of short-term plasticity, we examined the effects of inhibitors of protein kinases A (PKA) and C (PKC). While inhibition of PKC had little effect, PKA inhibition reduced augmentation by 76% and potentiation by 60%. Further, elevation of intracellular cAMP concentration, by either forskolin or IBMX, greatly increased mEPSC frequency and occluded the amount of augmentation and potentiation evoked by electrical stimulation. Finally, mutating a PKA phosphorylation site to non-phosphorylatable alanine largely abolished the ability of synapsin IIIa to rescue both augmentation and potentiation. Together, these results indicate that PKA activation is required for both augmentation and potentiation of spontaneous neurotransmitter release and that PKA-mediated phosphorylation of synapsin IIIa underlies both forms of presynaptic short-term plasticity.
Collapse
Affiliation(s)
- Qing Cheng
- Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, United States
| | - Sang-Ho Song
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - George J Augustine
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,Institute of Molecular and Cell Biology, Singapore, Singapore
| |
Collapse
|
11
|
Michetti C, Caruso A, Pagani M, Sabbioni M, Medrihan L, David G, Galbusera A, Morini M, Gozzi A, Benfenati F, Scattoni ML. The Knockout of Synapsin II in Mice Impairs Social Behavior and Functional Connectivity Generating an ASD-like Phenotype. Cereb Cortex 2018; 27:5014-5023. [PMID: 28922833 DOI: 10.1093/cercor/bhx207] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Indexed: 12/28/2022] Open
Abstract
Autism spectrum disorders (ASD) and epilepsy are neurodevelopmental conditions that appear with high rate of co-occurrence, suggesting the possibility of a common genetic basis. Mutations in Synapsin (SYN) genes, particularly SYN1 and SYN2, have been recently associated with ASD and epilepsy in humans. Accordingly, mice lacking Syn1 or Syn2, but not Syn3, experience epileptic seizures and display autistic-like traits that precede the onset of seizures. Here, we analyzed social behavior and ultrasonic vocalizations emitted in 2 social contexts by SynI, SynII, or SynIII mutants and show that SynII mutants display the most severe ASD-like phenotype. We also show that the behavioral SynII phenotype correlates with a significant decrease in auditory and hippocampal functional connectivity as measured with resting state functional magnetic resonance imaging (rsfMRI). Taken together, our results reveal a permissive contribution of Syn2 to the expression of normal socio-communicative behavior, and suggest that Syn2-mediated synaptic dysfunction can lead to ASD-like behavior through dysregulation of cortical connectivity.
Collapse
Affiliation(s)
- Caterina Michetti
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova 16132, Italy
| | - Angela Caruso
- Research Coordination and support Service, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Marco Pagani
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems @ UniTn, Istituto Italiano di Tecnologia, Rovereto 38068, Italy.,Center for Mind and Brain Sciences, University of Trento, Rovereto 38068, Italy
| | - Mara Sabbioni
- Research Coordination and support Service, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Lucian Medrihan
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova 16132, Italy
| | - Gergely David
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems @ UniTn, Istituto Italiano di Tecnologia, Rovereto 38068, Italy
| | - Alberto Galbusera
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems @ UniTn, Istituto Italiano di Tecnologia, Rovereto 38068, Italy
| | - Monica Morini
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova 16132, Italy
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems @ UniTn, Istituto Italiano di Tecnologia, Rovereto 38068, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova 16132, Italy
| | - Maria Luisa Scattoni
- Research Coordination and support Service, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
12
|
Barbieri R, Contestabile A, Ciardo MG, Forte N, Marte A, Baldelli P, Benfenati F, Onofri F. Synapsin I and Synapsin II regulate neurogenesis in the dentate gyrus of adult mice. Oncotarget 2018; 9:18760-18774. [PMID: 29721159 PMCID: PMC5922353 DOI: 10.18632/oncotarget.24655] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 02/25/2018] [Indexed: 01/23/2023] Open
Abstract
Adult neurogenesis is emerging as an important player in brain functions and homeostasis, while impaired or altered adult neurogenesis has been associated with a number of neuropsychiatric diseases, such as depression and epilepsy. Here we investigated the possibility that synapsins (Syns) I and II, beyond their known functions in developing and mature neurons, also play a role in adult neurogenesis. We performed a systematic evaluation of the distinct stages of neurogenesis in the hippocampal dentate gyrus of Syn I and Syn II knockout (KO) mice, before (2-months-old) and after (6-months-old) the appearance of the epileptic phenotype. We found that Syns I and II play an important role in the regulation of adult neurogenesis. In juvenile mice, Syn II deletion was associated with a specific decrease in the proliferation of neuronal progenitors, whereas Syn I deletion impaired the survival of newborn neurons. These defects were reverted after the appearance of the epileptic phenotype, with Syn I KO and Syn II KO mice exhibiting significant increases in survival and proliferation, respectively. Interestingly, long-term potentiation dependent on newborn neurons was present in both juvenile Syn mutants while, at later ages, it was only preserved in Syn II KO mice that also displayed an increased expression of brain-derived neurotrophic factor. This study suggests that Syns I and II play a role in adult neurogenesis and the defects in neurogenesis associated with Syn deletion may contribute to the alterations of cognitive functions observed in Syn-deficient mice.
Collapse
Affiliation(s)
- Raffaella Barbieri
- Department of Experimental Medicine, University of Genova, 16132, Genova, Italy
| | - Andrea Contestabile
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, 16163, Genova, Italy
| | - Maria Grazia Ciardo
- Department of Experimental Medicine, University of Genova, 16132, Genova, Italy
| | - Nicola Forte
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132, Genova, Italy
| | - Antonella Marte
- Department of Experimental Medicine, University of Genova, 16132, Genova, Italy
| | - Pietro Baldelli
- Department of Experimental Medicine, University of Genova, 16132, Genova, Italy.,Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132, Genova, Italy
| | - Fabio Benfenati
- Department of Experimental Medicine, University of Genova, 16132, Genova, Italy.,Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132, Genova, Italy
| | - Franco Onofri
- Department of Experimental Medicine, University of Genova, 16132, Genova, Italy
| |
Collapse
|
13
|
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: 15] [Impact Index Per Article: 2.5] [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
|
14
|
Hu Y, Pan S, Zhang HT. Interaction of Cdk5 and cAMP/PKA Signaling in the Mediation of Neuropsychiatric and Neurodegenerative Diseases. ADVANCES IN NEUROBIOLOGY 2018; 17:45-61. [PMID: 28956329 DOI: 10.1007/978-3-319-58811-7_3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Both cyclin-dependent kinase 5 (Cdk5) and cyclic AMP (cAMP)/protein kinase A (PKA) regulate fundamental central nervous system (CNS) functions including neuronal survival, neurite and axonal outgrowth, neuron development and cognition. Cdk5, a serine/threonine kinase, is activated by p35 or p39 and phosphorylates multiple signaling components of various pathways, including cAMP/PKA signaling. Here, we review the recent literature on the interaction between Cdk5 and cAMP/PKA signaling and their role in the mediation of CNS functions and neuropsychiatric and neurodegenerative diseases.
Collapse
Affiliation(s)
- Yafang Hu
- Department of Neurology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong, 510515, China.
| | - Suyue Pan
- Department of Neurology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong, 510515, China
| | - Han-Ting Zhang
- Department of Behavioral Medicine and Psychiatry, West Virginia University Health Sciences Center, 1 Medical Center Drive, Morgantown, WV, 26506, USA.,Department of Physiology and Pharmacology, West Virginia University Health Sciences Center, 1 Medical Center Drive, Morgantown, WV, 26506, USA.,Institute of Pharmacology, Taishan Medical University, Taian, 271016, China
| |
Collapse
|
15
|
Mirza FJ, Zahid S. The Role of Synapsins in Neurological Disorders. Neurosci Bull 2017; 34:349-358. [PMID: 29282612 DOI: 10.1007/s12264-017-0201-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 10/27/2017] [Indexed: 12/12/2022] Open
Abstract
Synapsins serve as flagships among the presynaptic proteins due to their abundance on synaptic vesicles and contribution to synaptic communication. Several studies have emphasized the importance of this multi-gene family of neuron-specific phosphoproteins in maintaining brain physiology. In the recent times, increasing evidence has established the relevance of alterations in synapsins as a major determinant in many neurological disorders. Here, we give a comprehensive description of the diverse roles of the synapsin family and the underlying molecular mechanisms that contribute to several neurological disorders. These physiologically important roles of synapsins associated with neurological disorders are just beginning to be understood. A detailed understanding of the diversified expression of synapsins may serve to strategize novel therapeutic approaches for these debilitating neurological disorders.
Collapse
Affiliation(s)
- Fatima Javed Mirza
- Neurobiology Research Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Saadia Zahid
- Neurobiology Research Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan.
| |
Collapse
|
16
|
Kao HT, Ryoo K, Lin A, Janoschka SR, Augustine GJ, Porton B. Synapsins regulate brain-derived neurotrophic factor-mediated synaptic potentiation and axon elongation by acting on membrane rafts. Eur J Neurosci 2017; 45:1085-1101. [PMID: 28245069 DOI: 10.1111/ejn.13552] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 01/27/2017] [Accepted: 02/15/2017] [Indexed: 11/29/2022]
Abstract
In neurons, intracellular membrane rafts are essential for specific actions of brain-derived neurotrophic factor (BDNF), which include the regulation of axon outgrowth, growth cone turning and synaptic transmission. Virtually, all the actions of BDNF are mediated by binding to its receptor, TrkB. The association of TrkB with the tyrosine kinase, Fyn, is critical for its localization to intracellular membrane rafts. Here, we show that synapsins, a family of highly amphipathic neuronal phosphoproteins, regulate membrane raft lipid composition and consequently, the ability of BDNF to regulate axon/neurite development and potentiate synaptic transmission. In the brains of mice lacking all synapsins, the expression of both BDNF and TrkB were increased, suggesting that BDNF/TrkB-mediated signaling is impaired. Consistent with this finding, synapsin-depleted neurons exhibit altered raft lipid composition, deficient targeting of Fyn to rafts, attenuated TrkB activation, and abrogation of BDNF-stimulated axon outgrowth and synaptic potentiation. Conversely, overexpression of synapsins in neuroblastoma cells results in corresponding reciprocal changes in raft lipid composition, increased localization of Fyn to rafts and promotion of BDNF-stimulated neurite formation. In the presence of synapsins, the ratio of cholesterol to estimated total phospholipids converged to 1, suggesting that synapsins act by regulating the ratio of lipids in intracellular membranes, thereby promoting lipid raft formation. These studies reveal a mechanistic link between BDNF and synapsins, impacting early development and synaptic transmission.
Collapse
Affiliation(s)
- Hung-Teh Kao
- Department of Psychiatry and Human Behavior, Brown University, 171 Meeting Street, Room 187, Providence, RI, 02912, USA.,Butler Hospital, Providence, RI, USA
| | - Kanghyun Ryoo
- Center for Functional Connectomics, Korea Institute of Science and Technology, Sungbukgu, Seoul, Korea
| | - Albert Lin
- Department of Psychiatry and Human Behavior, Brown University, 171 Meeting Street, Room 187, Providence, RI, 02912, USA.,Butler Hospital, Providence, RI, USA
| | - Stephen R Janoschka
- Department of Psychiatry and Human Behavior, Brown University, 171 Meeting Street, Room 187, Providence, RI, 02912, USA.,Butler Hospital, Providence, RI, USA
| | - George J Augustine
- Center for Functional Connectomics, Korea Institute of Science and Technology, Sungbukgu, Seoul, Korea.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Barbara Porton
- Department of Psychiatry and Human Behavior, Brown University, 171 Meeting Street, Room 187, Providence, RI, 02912, USA.,Butler Hospital, Providence, RI, USA
| |
Collapse
|
17
|
Gonzalez-Lozano MA, Klemmer P, Gebuis T, Hassan C, van Nierop P, van Kesteren RE, Smit AB, Li KW. Dynamics of the mouse brain cortical synaptic proteome during postnatal brain development. Sci Rep 2016; 6:35456. [PMID: 27748445 PMCID: PMC5066275 DOI: 10.1038/srep35456] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 09/28/2016] [Indexed: 01/04/2023] Open
Abstract
Development of the brain involves the formation and maturation of numerous synapses. This process requires prominent changes of the synaptic proteome and potentially involves thousands of different proteins at every synapse. To date the proteome analysis of synapse development has been studied sparsely. Here, we analyzed the cortical synaptic membrane proteome of juvenile postnatal days 9 (P9), P15, P21, P27, adolescent (P35) and different adult ages P70, P140 and P280 of C57Bl6/J mice. Using a quantitative proteomics workflow we quantified 1560 proteins of which 696 showed statistically significant differences over time. Synaptic proteins generally showed increased levels during maturation, whereas proteins involved in protein synthesis generally decreased in abundance. In several cases, proteins from a single functional molecular entity, e.g., subunits of the NMDA receptor, showed differences in their temporal regulation, which may reflect specific synaptic development features of connectivity, strength and plasticity. SNARE proteins, Snap 29/47 and Stx 7/8/12, showed higher expression in immature animals. Finally, we evaluated the function of Cxadr that showed high expression levels at P9 and a fast decline in expression during neuronal development. Knock down of the expression of Cxadr in cultured primary mouse neurons revealed a significant decrease in synapse density.
Collapse
Affiliation(s)
- Miguel A Gonzalez-Lozano
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Patricia Klemmer
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Titia Gebuis
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Chopie Hassan
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Pim van Nierop
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Ronald E van Kesteren
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Ka Wan Li
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics &Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Winkle CC, Taylor KL, Dent EW, Gallo G, Greif KF, Gupton SL. Beyond the cytoskeleton: The emerging role of organelles and membrane remodeling in the regulation of axon collateral branches. Dev Neurobiol 2016; 76:1293-1307. [PMID: 27112549 DOI: 10.1002/dneu.22398] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/11/2016] [Accepted: 04/21/2016] [Indexed: 12/19/2022]
Abstract
The generation of axon collateral branches is a fundamental aspect of the development of the nervous system and the response of axons to injury. Although much has been discovered about the signaling pathways and cytoskeletal dynamics underlying branching, additional aspects of the cell biology of axon branching have received less attention. This review summarizes recent advances in our understanding of key factors involved in axon branching. This article focuses on how cytoskeletal mechanisms, intracellular organelles, such as mitochondria and the endoplasmic reticulum, and membrane remodeling (exocytosis and endocytosis) contribute to branch initiation and formation. Together this growing literature provides valuable insight as well as a platform for continued investigation into how multiple aspects of axonal cell biology are spatially and temporally orchestrated to give rise to axon branches. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 76: 1293-1307, 2016.
Collapse
Affiliation(s)
- Cortney C Winkle
- Neurobiology Curriculum, University of North Carolina, Chapel Hill, North Carolina, 27599
| | - Kendra L Taylor
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, Wisconsin, 53705
| | - Erik W Dent
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, 53705
| | - Gianluca Gallo
- Lewis Katz School of Medicine, Department of Anatomy and Cell Biology, Shriners Hospitals Pediatric Research Center, Temple University, Philadelphia, Pennsylvania, 19140
| | - Karen F Greif
- Department of Biology, Bryn Mawr College, Bryn Mawr, Pennsylvania, 19010
| | - Stephanie L Gupton
- Department of Cell Biology and Physiology, Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina, 27599
| |
Collapse
|
19
|
Marte A, Messa M, Benfenati F, Onofri F. Synapsins Are Downstream Players of the BDNF-Mediated Axonal Growth. Mol Neurobiol 2016; 54:484-494. [PMID: 26742525 DOI: 10.1007/s12035-015-9659-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 12/17/2015] [Indexed: 01/09/2023]
Abstract
Synapsins (Syns) are synaptic vesicle-associated phosphoproteins involved in neuronal development and neurotransmitter release. While Syns are implicated in the regulation of brain-derived neurotrophic factor (BDNF)-induced neurotransmitter release, their role in the BDNF developmental effects has not been fully elucidated. By using primary cortical neurons from Syn I knockout (KO) and Syn I/II/III KO mice, we studied the effects of BDNF and nerve growth factor (NGF) on axonal growth. While NGF had similar effects in all genotypes, BDNF induced significant differences in Syn KO axonal outgrowth compared to wild type (WT), an effect that was rescued by the re-expression of Syn I. Moreover, the significant increase of axonal branching induced by BDNF in WT neurons was not detectable in Syn KO neurons. The expression analysis of BDNF receptors in Syn KO neurons revealed a significant decrease of the full length TrkB receptor and an increase in the levels of the truncated TrkB.t1 isoform and p75NTR associated with a marked reduction of the BDNF-induced MAPK/Erk activation. By using the Trk inhibitor K252a, we demonstrated that these differences in BDNF effects were dependent on a TrkB/p75NTR imbalance. The data indicate that Syn I plays a pivotal role in the BDNF signal transduction during axonal growth.
Collapse
Affiliation(s)
- Antonella Marte
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, 16132, Genova, Italy
| | - Mirko Messa
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Cell Biology, Howard Hughes Medical Institute, Yale University School of Medicine, 295 Congress Avenue, 06519, New Haven, CT, USA
| | - Fabio Benfenati
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, 16132, Genova, Italy
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genova, Italy
| | - Franco Onofri
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, 16132, Genova, Italy.
| |
Collapse
|
20
|
Nikolaev M, Heggelund P. Functions of synapsins in corticothalamic facilitation: important roles of synapsin I. J Physiol 2015; 593:4499-510. [PMID: 26256545 DOI: 10.1113/jp270553] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 08/04/2015] [Indexed: 12/15/2022] Open
Abstract
KEY POINTS The synaptic vesicle associated proteins synapsin I and synapsin II have important functions in synaptic short-term plasticity. We investigated their functions in cortical facilitatory feedback to neurons in dorsal lateral geniculate nucleus (dLGN), feedback that has important functions in state-dependent regulation of thalamic transmission of visual input to cortex. We compared results from normal wild-type (WT) mice and synapsin knockout (KO) mice in several types of synaptic plasticity, and found clear differences between the responses of neurons in the synapsin I KO and the WT, but no significant differences between the synapsin II KO and the WT. These results are in contrast to the important role of synapsin II previously demonstrated in similar types of synaptic plasticity in other brain regions, indicating that the synapsins can have different roles in similar types of STP in different parts of the brain. ABSTRACT The synaptic vesicle associated proteins synapsin I (SynI) and synapsin II (SynII) have important functions in several types of synaptic short-term plasticity in the brain, but their separate functions in different types of synapses are not well known. We investigated possible distinct functions of the two synapsins in synaptic short-term plasticity at corticothalamic synapses on relay neurons in the dorsal lateral geniculate nucleus. These synapses provide excitatory feedback from visual cortex to the relay cells, feedback that can facilitate transmission of signals from retina to cortex. We compared results from normal wild-type (WT), SynI knockout (KO) and SynII KO mice, in three types of synaptic plasticity mainly linked to presynaptic mechanism. In SynI KO mice, paired-pulse stimulation elicited increased facilitation at short interpulse intervals compared to the WT. Pulse-train stimulation elicited weaker facilitation than in the WT, and also post-tetanic potentiation was weaker in SynI KO than in the WT. Between SynII KO and the WT we found no significant differences. Thus, SynI has important functions in these types of synaptic plasticity at corticothalamic synapses. Interestingly, our data are in contrast to the important role of SynII previously shown for sustained synaptic transmission during intense stimulation in excitatory synapses in other parts of the brain, and our results suggest that SynI and SynII may have different roles in similar types of STP in different parts of the brain.
Collapse
Affiliation(s)
- Maxim Nikolaev
- Institute of Basic Medical Sciences, University of Oslo, N-0317, Oslo, Norway.,I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, 194223, 44 Thorez pr., St Petersburg, Russia
| | - Paul Heggelund
- Institute of Basic Medical Sciences, University of Oslo, N-0317, Oslo, Norway
| |
Collapse
|
21
|
Brenes O, Giachello CNG, Corradi AM, Ghirardi M, Montarolo PG. Synapsin knockdown is associated with decreased neurite outgrowth, functional synaptogenesis impairment, and fast high-frequency neurotransmitter release. J Neurosci Res 2015. [PMID: 26213348 DOI: 10.1002/jnr.23624] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Synapsins (Syns) are an evolutionarily conserved family of synaptic vesicle-associated proteins related to fine tuning of synaptic transmission. Studies with mammals have partially clarified the different roles of Syns; however, the presence of different genes and isoforms and the development of compensatory mechanisms hinder accurate data interpretation. Here, we use a simple in vitro monosynaptic Helix neuron connection, reproducing an in vivo physiological connection as a reliable experimental model to investigate the effects of Syn knockdown. Cells overexpressing an antisense construct against Helix Syn showed a time-dependent decrease of Syn immunostaining, confirming protein loss. At the morphological level, Syn-silenced cells showed a reduction in neurite linear outgrowth and branching and in the size and number of synaptic varicosities. Functionally, Syn-silenced cells presented a reduced ability to form synaptic connections; however, functional chemical synapses showed similar basal excitatory postsynaptic potentials and similar short-term plasticity paradigms. In addition, Syn-silenced cells presented faster neurotransmitter release and decreased postsynaptic response toward the end of long tetanic presynaptic stimulations, probably related to an impairment of the synaptic vesicle trafficking resulting from a different vesicle handling, with an increased readily releasable pool and a compromised reserve pool.
Collapse
Affiliation(s)
- Oscar Brenes
- Department of Neuroscience, Section of Physiology, University of Turin, Turin, Italy.,Department of Physiology, School of Medicine, University of Costa Rica, San José, Costa Rica
| | | | | | - Mirella Ghirardi
- Department of Neuroscience, Section of Physiology, University of Turin, Turin, Italy.,National Institute of Neuroscience, Turin, Italy
| | - Pier Giorgio Montarolo
- Department of Neuroscience, Section of Physiology, University of Turin, Turin, Italy.,National Institute of Neuroscience, Turin, Italy
| |
Collapse
|
22
|
Perlini LE, Szczurkowska J, Ballif BA, Piccini A, Sacchetti S, Giovedì S, Benfenati F, Cancedda L. Synapsin III acts downstream of semaphorin 3A/CDK5 signaling to regulate radial migration and orientation of pyramidal neurons in vivo. Cell Rep 2015; 11:234-48. [PMID: 25843720 PMCID: PMC4405008 DOI: 10.1016/j.celrep.2015.03.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 02/13/2015] [Accepted: 03/06/2015] [Indexed: 11/04/2022] Open
Abstract
Synapsin III (SynIII) is a phosphoprotein that is highly expressed at early stages of neuronal development. Whereas in vitro evidence suggests a role for SynIII in neuronal differentiation, in vivo evidence is lacking. Here, we demonstrate that in vivo downregulation of SynIII expression affects neuronal migration and orientation. By contrast, SynIII overexpression affects neuronal migration, but not orientation. We identify a cyclin-dependent kinase-5 (CDK5) phosphorylation site on SynIII and use phosphomutant rescue experiments to demonstrate its role in SynIII function. Finally, we show that SynIII phosphorylation at the CDK5 site is induced by activation of the semaphorin-3A (Sema3A) pathway, which is implicated in migration and orientation of cortical pyramidal neurons (PNs) and is known to activate CDK5. Thus, fine-tuning of SynIII expression and phosphorylation by CDK5 activation through Sema3A activity is essential for proper neuronal migration and orientation. Precise regulation of SynIII expression is essential during brain development SynIII regulates neuronal migration, orientation, and morphological maturation SynIII acts downstream of the Sema3A pathway, which involves NP1 and kinase CDK5 Phosphorylation of SynIII by CDK5 on Ser404 is essential for SynIII function
Collapse
Affiliation(s)
- Laura E Perlini
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa 16163, Italy; Department of Experimental Medicine, University of Genoa, Genoa 16132, Italy
| | - Joanna Szczurkowska
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa 16163, Italy
| | - Bryan A Ballif
- Department of Biology, University of Vermont, Burlington, VT 05405-0086, USA
| | - Alessandra Piccini
- Department of Experimental Medicine, University of Genoa, Genoa 16132, Italy
| | - Silvio Sacchetti
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa 16163, Italy
| | - Silvia Giovedì
- Department of Experimental Medicine, University of Genoa, Genoa 16132, Italy
| | - Fabio Benfenati
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa 16163, Italy; Department of Experimental Medicine, University of Genoa, Genoa 16132, Italy
| | - Laura Cancedda
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa 16163, Italy.
| |
Collapse
|
23
|
Garbarino G, Costa S, Pestarino M, Candiani S. Differential expression of synapsin genes during early zebrafish development. Neuroscience 2014; 280:351-67. [DOI: 10.1016/j.neuroscience.2014.09.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 09/03/2014] [Accepted: 09/03/2014] [Indexed: 11/27/2022]
|
24
|
Yu WJ, Li NN, Tan EK, Cheng L, Zhang JH, Mao XY, Chang XL, Zhao DM, Liao Q, Peng R. No association of four candidate genetic variants in MnSOD and SYNIII with Parkinson's disease in two Chinese populations. PLoS One 2014; 9:e88050. [PMID: 24586301 PMCID: PMC3935830 DOI: 10.1371/journal.pone.0088050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 01/02/2014] [Indexed: 02/05/2023] Open
Abstract
Background The manganese superoxide dismutase (MnSOD) gene, which encodes a chief reactive oxygen species (ROS) scavenging enzyme, has been reported to be associated with the risk of developing sporadic Parkinson's disease (PD) in some Asian races and the synapsin III (SYN3) gene with some neuropsychiatric diseases. Objective: To explore the associations between the MnSOD and SYN III variations and PD in two Chinese populations from mainland China and Singapore. Methods We recruited 2342 subjects including 1200 sporadic PD patients and 1142 healthy controls from two independent Asian countries. Using a case-control methodology, we genotyped the single nucleotide polymorphisms (SNP) in MnSOD (rs4880) and SYN III (rs3788470, rs3827336, rs5998557) to explore the associations with risk of PD. Results The results showed the genotype distributions and minor allele frequencies (MAF) of MnSOD (rs4880) and SYN III (rs3788470, rs3827336, rs5998557) were not significantly different between PD patients and healthy controls in mainland China and Singapore, as well as in merged populations. Conclusions The variations of MnSOD (rs4880) and SYN III (rs3788470, rs3827336, rs5998557) were not major risk factors for PD among Chinese, at least in our study populations.
Collapse
Affiliation(s)
- Wen Juan Yu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Nan Nan Li
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Eng King Tan
- Duke–NUS Graduate Medical School, Singapore, Singapore
- Department of Neurology, Singapore General Hospital, National Neuroscience Institute, Singapore, Singapore
| | - Lan Cheng
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Jin Hong Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, PR China
- Department of Internal Medicine, Wangjiang Hospital, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Xue Ye Mao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Xue Li Chang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Dong Mei Zhao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Qiao Liao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Rong Peng
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, PR China
- * E-mail:
| |
Collapse
|
25
|
Giovedí S, Corradi A, Fassio A, Benfenati F. Involvement of synaptic genes in the pathogenesis of autism spectrum disorders: the case of synapsins. Front Pediatr 2014; 2:94. [PMID: 25237665 PMCID: PMC4154395 DOI: 10.3389/fped.2014.00094] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 08/21/2014] [Indexed: 12/03/2022] Open
Abstract
Autism spectrum disorders (ASDs) are heterogeneous neurodevelopmental disorders characterized by deficits in social interaction and social communication, restricted interests, and repetitive behaviors. Many synaptic protein genes are linked to the pathogenesis of ASDs, making them prototypical synaptopathies. An array of mutations in the synapsin (Syn) genes in humans has been recently associated with ASD and epilepsy, diseases that display a frequent comorbidity. Syns are pre-synaptic proteins regulating synaptic vesicle traffic, neurotransmitter release, and short-term synaptic plasticity. In doing so, Syn isoforms control the tone of activity of neural circuits and the balance between excitation and inhibition. As ASD pathogenesis is believed to result from dysfunctions in the balance between excitatory and inhibitory transmissions in neocortical areas, Syns are novel ASD candidate genes. Accordingly, deletion of single Syn genes in mice, in addition to epilepsy, causes core symptoms of ASD by affecting social behavior, social communication, and repetitive behaviors. Thus, Syn knockout mice represent a good experimental model to define synaptic alterations involved in the pathogenesis of ASD and epilepsy.
Collapse
Affiliation(s)
- Silvia Giovedí
- Department of Experimental Medicine, University of Genova , Genova , Italy
| | - Anna Corradi
- Department of Experimental Medicine, University of Genova , Genova , Italy
| | - Anna Fassio
- Department of Experimental Medicine, University of Genova , Genova , Italy ; Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia , Genova , Italy
| | - Fabio Benfenati
- Department of Experimental Medicine, University of Genova , Genova , Italy ; Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia , Genova , Italy
| |
Collapse
|
26
|
Bas E, Van De Water TR, Lumbreras V, Rajguru S, Goss G, Hare JM, Goldstein BJ. Adult human nasal mesenchymal-like stem cells restore cochlear spiral ganglion neurons after experimental lesion. Stem Cells Dev 2013; 23:502-14. [PMID: 24172073 DOI: 10.1089/scd.2013.0274] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A loss of sensory hair cells or spiral ganglion neurons from the inner ear causes deafness, affecting millions of people. Currently, there is no effective therapy to repair the inner ear sensory structures in humans. Cochlear implantation can restore input, but only if auditory neurons remain intact. Efforts to develop stem cell-based treatments for deafness have demonstrated progress, most notably utilizing embryonic-derived cells. In an effort to bypass limitations of embryonic or induced pluripotent stem cells that may impede the translation to clinical applications, we sought to utilize an alternative cell source. Here, we show that adult human mesenchymal-like stem cells (MSCs) obtained from nasal tissue can repair spiral ganglion loss in experimentally lesioned cochlear cultures from neonatal rats. Stem cells engraft into gentamicin-lesioned organotypic cultures and orchestrate the restoration of the spiral ganglion neuronal population, involving both direct neuronal differentiation and secondary effects on endogenous cells. As a physiologic assay, nasal MSC-derived cells engrafted into lesioned spiral ganglia demonstrate responses to infrared laser stimulus that are consistent with those typical of excitable cells. The addition of a pharmacologic activator of the canonical Wnt/β-catenin pathway concurrent with stem cell treatment promoted robust neuronal differentiation. The availability of an effective adult autologous cell source for inner ear tissue repair should contribute to efforts to translate cell-based strategies to the clinic.
Collapse
Affiliation(s)
- Esperanza Bas
- 1 Department of Otolaryngology, University of Miami Miller School of Medicine , Miami, Florida
| | | | | | | | | | | | | |
Collapse
|
27
|
Owe SG, Erisir A, Heggelund P. Terminals of the major thalamic input to visual cortex are devoid of synapsin proteins. Neuroscience 2013; 243:115-25. [PMID: 23535254 DOI: 10.1016/j.neuroscience.2013.03.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 02/12/2013] [Accepted: 03/13/2013] [Indexed: 01/13/2023]
Abstract
Synapsins are nerve-terminal proteins that are linked to synaptic transmission and key factors in several forms of synaptic plasticity. While synapsins are generally assumed to be ubiquitous in synaptic terminals, whether they are excluded from certain types of terminals is of interest. In the visual pathway, synapsins are lacking in photoreceptor and bipolar cell terminals as well as in retinogeniculate synapses. These are the terminals of the first three feedforward synapses in the visual pathway, implying that lack of synapsins may be a common property of terminals that provide the primary driver activity onto their postsynaptic neurons. To further investigate this idea, we studied the fourth driver synapse, thalamocortical synapses in visual cortex, using glutamatergic terminal antibody markers anti-VGluT1 and VGluT2, anti-Synapsin I and II, and confocal microscopy to analyze co-localization of these proteins in terminals. We also used pre-embedding immunocytochemical labeling followed by electron microscopy to investigate morphological similarities or differences between terminals containing synapsins or VGluT2. In visual cortex, synapsin coincided extensively with non-TC-neuron marker, VGluT1, while thalamocortical terminal marker VGluT2 and synapsin overlap was sparse. Morphologically, synapsin-stained terminals were smaller than non-stained, while VGluT2-positive thalamocortical terminals constituted the largest terminals in cortex. The size discrepancy between synapsin- and VGluT2-positive terminals, together with the complementary staining patterns, indicates that thalamocortical synapses are devoid of synapsins, and support the hypothesis that afferent sensory information is consistently transmitted without the involvement of synapsins. Furthermore, VGluT2 and synapsins were colocalized in other brain structures, suggesting that lack of synapsins is not a property of VGluT2-containing terminals, but a property of primary driver terminals in the visual system.
Collapse
Affiliation(s)
- S G Owe
- Institute of Basic Medical Sciences, Department of Physiology, University of Oslo, N-0317 Oslo, Norway
| | | | | |
Collapse
|
28
|
H3K4 tri-methylation in synapsin genes leads to different expression patterns in bipolar disorder and major depression. Int J Neuropsychopharmacol 2013; 16:289-99. [PMID: 22571925 PMCID: PMC3564952 DOI: 10.1017/s1461145712000363] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The synapsin family of neuronal phosphoproteins is composed of three genes (SYN1, SYN2 and SYN3) with alternative splicing resulting in a number of variants with various levels of homology. These genes have been postulated to play significant roles in several neuropsychiatric disorders, including bipolar disorder, schizophrenia and epilepsy. Epigenetic regulatory mechanisms, such as histone modifications in gene regulatory regions, have also been proposed to play a role in a number of psychiatric disorders, including bipolar disorder and major depressive disorder. One of the best characterized histone modifications is histone 3 lysine 4 tri-methylation (H3K4me3), an epigenetic mark shown to be highly enriched at transcriptional start sites and associated with active transcription. In the present study we have quantified the expression of transcript variants of the three synapsin genes and investigated their relationship to H3K4me3 promoter enrichment in post-mortem brain samples. We found that histone modification marks were significantly increased in bipolar disorder and major depression and this effect was correlated with significant increases in gene expression. Our findings suggest that synapsin dysregulation in mood disorders is mediated in part by epigenetic regulatory mechanisms.
Collapse
|
29
|
Greco B, Managò F, Tucci V, Kao HT, Valtorta F, Benfenati F. Autism-related behavioral abnormalities in synapsin knockout mice. Behav Brain Res 2012; 251:65-74. [PMID: 23280234 PMCID: PMC3730181 DOI: 10.1016/j.bbr.2012.12.015] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 11/19/2012] [Accepted: 12/10/2012] [Indexed: 02/08/2023]
Abstract
Several synaptic genes predisposing to autism-spectrum disorder (ASD) have been identified. Nonsense and missense mutations in the SYN1 gene encoding for Synapsin I have been identified in families segregating for idiopathic epilepsy and ASD and genetic mapping analyses have identified variations in the SYN2 gene as significantly contributing to epilepsy predisposition. Synapsins (Syn I/II/III) are a multigene family of synaptic vesicle-associated phosphoproteins playing multiple roles in synaptic development, transmission and plasticity. Lack of SynI and/or SynII triggers a strong epileptic phenotype in mice associated with mild cognitive impairments that are also present in the non-epileptic SynIII(-/-) mice. SynII(-/-) and SynIII(-/-) mice also display schizophrenia-like traits, suggesting that Syns could be involved in the regulation of social behavior. Here, we studied social interaction and novelty, social recognition and social dominance, social transmission of food preference and social memory in groups of male SynI(-/-), SynII(-/-) and SynIII(-/-) mice before and after the appearance of the epileptic phenotype and compared their performances with control mice. We found that deletion of Syn isoforms widely impairs social behaviors and repetitive behaviors, resulting in ASD-related phenotypes. SynI or SynIII deletion altered social behavior, whereas SynII deletion extensively impaired various aspects of social behavior and memory, altered exploration of a novel environment and increased self-grooming. Social impairments of SynI(-/-) and SynII(-/-) mice were evident also before the onset of seizures. The results demonstrate an involvement of Syns in generation of the behavioral traits of ASD and identify Syn knockout mice as a useful experimental model of ASD and epilepsy.
Collapse
Affiliation(s)
- Barbara Greco
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova, Italy
| | | | | | | | | | | |
Collapse
|
30
|
Gene expression profile analysis in cultured human neuronal cells after static magnetic stimulation. BIOCHIP JOURNAL 2012. [DOI: 10.1007/s13206-012-6308-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
31
|
Temporal evolution of neurophysiological and behavioral features of synapsin I/II/III triple knock-out mice. Epilepsy Res 2012; 103:153-60. [PMID: 22846639 PMCID: PMC3574234 DOI: 10.1016/j.eplepsyres.2012.07.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2012] [Revised: 06/07/2012] [Accepted: 07/10/2012] [Indexed: 11/23/2022]
Abstract
Deletion of one or more synapsin genes in mice results in a spontaneous epilepsy. In these animals, seizures can be evoked by opening or moving the cage. Aim of the present study was to characterize the evolution of the epileptic phenotype by neurophysiological examination and behavioral observation in synapsin triple knock-out (Syn-TKO) mice. Syn-TKO mice were studied from 20 postnatal days (PND) up to 6 months of age by video-EEG recording and behavioral observation. Background EEG spectral analysis was performed and data were compared to WT animals. Syn-TKO revealed rare spontaneous seizures and increased susceptibility to evoked seizures in mice from 60 to 100 PND. Spontaneous and evoked seizures presented similar duration and morphology. At times, seizures were followed by a post-ictal phase characterized by a 4 Hz rhythmic activity and immobility of the animal. Spectral analysis of background EEG evidenced a slowing of the theta-alpha peak in Syn-TKO mice compared to WT mice within the period from PND 40 to 100. These data indicate that Syn-TKO mice do not exhibit a linear progression of the epileptic phenotype, with the period corresponding to a higher susceptibility to evoked seizures characterized by background EEG slowing. This aspect might be connected to brain dysfunction often associated to epilepsy in the interictal period.
Collapse
|
32
|
Synaptic functions of invertebrate varicosities: what molecular mechanisms lie beneath. Neural Plast 2012; 2012:670821. [PMID: 22655209 PMCID: PMC3359714 DOI: 10.1155/2012/670821] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 02/27/2012] [Indexed: 11/26/2022] Open
Abstract
In mammalian brain, the cellular and molecular events occurring in both synapse formation and plasticity are difficult to study due to the large number of factors involved in these processes and because the contribution of each component is not well defined. Invertebrates, such as Drosophila, Aplysia, Helix, Lymnaea, and Helisoma, have proven to be useful models for studying synaptic assembly and elementary forms of learning. Simple nervous system, cellular accessibility, and genetic simplicity are some examples of the invertebrate advantages that allowed to improve our knowledge about evolutionary neuronal conserved mechanisms. In this paper, we present an overview of progresses that elucidates cellular and molecular mechanisms underlying synaptogenesis and synapse plasticity in invertebrate varicosities and their validation in vertebrates. In particular, the role of invertebrate synapsin in the formation of presynaptic terminals and the cell-to-cell interactions that induce specific structural and functional changes in their respective targets will be analyzed.
Collapse
|
33
|
Schock SC, Jolin-Dahel KS, Schock PC, Theiss S, Arbuthnott GW, Garcia-Munoz M, Staines WA. Development of dissociated cryopreserved rat cortical neurons in vitro. J Neurosci Methods 2012; 205:324-33. [PMID: 22326618 DOI: 10.1016/j.jneumeth.2012.01.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 01/25/2012] [Indexed: 11/30/2022]
Abstract
Dissociated neuronal cultures of various brain regions are commonly used to study physiological and pathophysiological processes in vitro. The data derived from these studies are often viewed to have relevance to processes taking place in the mature brain. However, due to the practical challenges associated with lengthy neuronal culture, neurons are often kept for 14 days in vitro (DIV), or less, before being subject to experimentation. Non-proliferative cultures such as primary neuronal cultures can be maintained for more than 42 DIV if water evaporation from culture media is monitored and corrected. To determine appropriate time points corresponding to the stages of cortical development, we compared characteristics of cryopreserved cortical neurons in cultures at various DIV using immunofluorescence, biochemical measurements and multielectrode array recordings. Compared to 21 and 35 DIV, at 14 DIV, cultures are still undergoing developmental changes and are not representative of adult in vivo brain tissue. Specifically, we noted significant lack in immunoreactivity for synaptic markers such as synapsin, vesicular GABA transporter and vesicular glutamate transporter at 14 DIV, relative to 21 and 35 DIV. Moreover, multielectrode array analysis indicated an increase in network firing up to 46 DIV with patterned firing peaking at 35 DIV. Our results provide specific evidence of the maturational stages of neurons in culture that can be used to more successfully plan various types of in vitro experimentation.
Collapse
Affiliation(s)
- Sarah C Schock
- Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | | | | | | | | | | | | |
Collapse
|
34
|
Porton B, Wetsel WC, Kao HT. Synapsin III: role in neuronal plasticity and disease. Semin Cell Dev Biol 2011; 22:416-24. [PMID: 21827867 DOI: 10.1016/j.semcdb.2011.07.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 07/13/2011] [Indexed: 12/31/2022]
Abstract
Synapsin III was discovered in 1998, more than two decades after the first two synapsins (synapsins I and II) were identified. Although the biology of synapsin III is not as well understood as synapsins I and II, this gene is emerging as an important factor in the regulation of the early stages of neurodevelopment and dopaminergic neurotransmission, and in certain neuropsychiatric illnesses. Molecular genetic and clinical studies of synapsin III have determined that its neurodevelopmental effects are exerted at the levels of neurogenesis and axonogenesis. In vitro voltammetry studies have shown that synapsin III can control dopamine release in the striatum. Since dopaminergic dysfunction is implicated in many neuropsychiatric conditions, one may anticipate that polymorphisms in synapsin III can exert pervasive effects, especially since it is localized to extrasynaptic sites. Indeed, mutations in this gene have been identified in individuals diagnosed with schizophrenia, bipolar disorder and multiple sclerosis. These and other findings indicate that the roles of synapsin III differ significantly from those of synapsins I and II. Here, we focus on the unique roles of the newest synapsin, and where relevant, compare and contrast these with the actions of synapsins I and II.
Collapse
Affiliation(s)
- Barbara Porton
- Department of Psychiatry and Human Behavior, Brown University, BioMedical Center, 171 Meeting Street, Room 187, Providence, RI 02912, USA
| | | | | |
Collapse
|
35
|
Fassio A, Raimondi A, Lignani G, Benfenati F, Baldelli P. Synapsins: from synapse to network hyperexcitability and epilepsy. Semin Cell Dev Biol 2011; 22:408-15. [PMID: 21816229 DOI: 10.1016/j.semcdb.2011.07.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 07/13/2011] [Indexed: 01/31/2023]
Abstract
The synapsin family in mammals consists of at least 10 isoforms encoded by three distinct genes and composed by a mosaic of conserved and variable domains. Synapsins, although not essential for the basic development and functioning of neuronal networks, are extremely important for the fine-tuning of SV cycling and neuronal plasticity. Single, double and triple synapsin knockout mice, with the notable exception of the synapsin III knockout mice, show a severe epileptic phenotype without gross alterations in brain morphology and connectivity. However, the molecular and physiological mechanisms underlying the pathogenesis of the epileptic phenotype observed in synapsin deficient mice are still far from being elucidated. In this review, we summarize the current knowledge about the role of synapsins in the regulation of network excitability and about the molecular mechanism leading to epileptic phenotype in mouse lines lacking one or more synapsin isoforms. The current evidences indicate that synapsins exert distinct roles in excitatory versus inhibitory synapses by differentially affecting crucial steps of presynaptic physiology and by this mean participate in the determination of network hyperexcitability.
Collapse
Affiliation(s)
- Anna Fassio
- Department of Experimental Medicine, Section of Physiology and National Institute of Neuroscience, University of Genova, Genova, Italy
| | | | | | | | | |
Collapse
|
36
|
Valtorta F, Pozzi D, Benfenati F, Fornasiero EF. The synapsins: multitask modulators of neuronal development. Semin Cell Dev Biol 2011; 22:378-86. [PMID: 21798361 DOI: 10.1016/j.semcdb.2011.07.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 07/13/2011] [Indexed: 01/10/2023]
Abstract
Neurons are examples of specialized cells that evolved the extraordinary ability to transmit electrochemical information in complex networks of interconnected cells. During their development, neurons undergo precisely regulated processes that define their lineage, positioning, morphogenesis and pattern of activity. The events leading to the establishment of functional neuronal networks follow a number of key steps, including asymmetric cell division from neuronal precursors, migration, establishment of polarity, neurite outgrowth and synaptogenesis. Synapsins are a family of abundant neuronal phosphoproteins that have been extensively studied for their role in the regulation of neurotransmission in presynaptic terminals. Beside their implication in the homeostasis of adult cells, synapsins influence the development of young neurons, interacting with cytoskeletal and vesicular components and regulating their dynamics. Although the exact molecular mechanisms determining synapsin function in neuronal development are still largely unknown, in this review we summarize the most important literature on the subject, providing a conceptual framework for the progress of present and future research.
Collapse
Affiliation(s)
- Flavia Valtorta
- San Raffaele Scientific Institute and Vita-Salute University, Via Olgettina 58, Milano, Italy.
| | | | | | | |
Collapse
|
37
|
Dagyte G, Luiten PG, De Jager T, Gabriel C, Mocaër E, Den Boer JA, Van der Zee EA. Chronic stress and antidepressant agomelatine induce region-specific changes in synapsin I expression in the rat brain. J Neurosci Res 2011; 89:1646-57. [PMID: 21688292 DOI: 10.1002/jnr.22697] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 04/18/2011] [Accepted: 04/18/2011] [Indexed: 12/17/2022]
Abstract
The antidepressant agomelatine acts as a melatonergic receptor (MT(1)/MT(2)) agonist and 5-HT(2C) receptor antagonist. Agomelatine has demonstrated efficacy in treating depression, but its neurobiological effects merit further investigation. Preclinical studies reported that agomelatine enhances adult hippocampal neurogenesis and increases expression of several neuroplasticity-associated molecules. Recently, we showed that agomelatine normalizes hippocampal neuronal activity and promotes neurogenesis in the stress-compromised brain. To characterize further the effects of this antidepressant in the stressed brain, here we investigated whether it induces changes in the expression of synapsin I (SynI), a regulator of synaptic transmission and plasticity. Adult male rats were subjected to daily footshock stress and agomelatine treatment for 3 weeks. Their brains were subsequently stained for total and phosphorylated SynI. Chronic footshock and agomelatine induced region-specific changes in SynI expression. Whereas chronic stress increased total SynI expression in all layers of the medial prefrontal cortex, agomelatine treatment abolished some of these effects. Furthermore, chronic agomelatine administration decreased total SynI expression in the hippocampal subregions of both stressed and nonstressed rats. Importantly, chronic stress decreased the fraction of phosphorylated SynI in all layers of the medial prefrontal cortex as well as selectively in the outer and middle molecular layers of the hippocampal dentate gyrus. These stress effects were at least partially abolished by agomelatine. Altogether, our data show that chronic stress and agomelatine treatment induce region-specific changes in SynI expression and its phosphorylation. Moreover, agomelatine partially counteracts the stress effects on SynI, suggesting a modulation of synaptic function by this antidepressant.
Collapse
Affiliation(s)
- Girstaute Dagyte
- Department of Molecular Neurobiology, University of Groningen, Groningen, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
38
|
Humeau Y, Candiani S, Ghirardi M, Poulain B, Montarolo P. Functional roles of synapsin: Lessons from invertebrates. Semin Cell Dev Biol 2011; 22:425-33. [DOI: 10.1016/j.semcdb.2011.07.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Accepted: 07/13/2011] [Indexed: 12/18/2022]
|
39
|
Compensatory network alterations upon onset of epilepsy in synapsin triple knock-out mice. Neuroscience 2011; 189:108-22. [PMID: 21621590 DOI: 10.1016/j.neuroscience.2011.05.030] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Revised: 04/16/2011] [Accepted: 05/11/2011] [Indexed: 11/21/2022]
Abstract
Adult synapsin triple-knockout mice exhibit epilepsy that manifests as generalized tonic-clonic seizures. Because in vitro recordings have shown a reduction in quantal release from inhibitory neurons, an inherent excitation-inhibition imbalance has been hypothesized as the direct culprit for epilepsy in these mice. We critically assessed this hypothesis by examining neurotransmission during the emergence of epilepsy. Using long-term video and telemetric EEG monitoring we found that synapsin triple-knockout mice exhibit an abrupt transition during early adulthood from a seizure-free presymptomatic latent state to a consistent symptomatic state of sensory-induced seizures. Electrophysiological recordings showed that during the latent period larger field responses could be elicited in slices from mutant mice. However, only after the transition to a symptomatic state in the adult mice did evoked epileptiform activity become prevalent. This state was characterized by resistance to the epileptiform-promoting effects of 4-aminopyridine, by marked hypersensitivity to blockage of GABAA receptors, and by the emergence of unresponsiveness to NMDA receptor antagonism, all of which were not observed during the latent period. Importantly, enhancement in inhibitory transmission was associated with upregulation of GAD67 expression without affecting the number of inhibitory neurons in the same brain areas where epileptiform activity was recorded. We therefore suggest that while deletion of the synapsins initially increases cortical network activity, this enhanced excitability is insufficient to elicit seizures. Rather, compensatory epileptogenic mechanisms are activated during the latent period that lead to an additional almost-balanced enhancement of both the excitatory and inhibitory components of the network, finally culminating in the emergence of epilepsy.
Collapse
|
40
|
Bloom OE, Morgan JR. Membrane trafficking events underlying axon repair, growth, and regeneration. Mol Cell Neurosci 2011; 48:339-48. [PMID: 21539917 DOI: 10.1016/j.mcn.2011.04.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 04/11/2011] [Accepted: 04/14/2011] [Indexed: 12/31/2022] Open
Abstract
Two central challenges for the field of neurobiology are to understand how axons grow and make proper synaptic connections under normal conditions and how they repair their membranes and mount regenerative responses after injury. At the most reductionist level, the first step toward addressing these challenges is to delineate the cellular and molecular processes by which an axon extends from its cell body. Underlying axon extension are questions of appropriate timing and mechanisms that establish or maintain the axon's polarity, initiate growth cone formation, and promote axon outgrowth and synapse formation. After injury, the problem is even more complicated because the neuron must also repair its damaged membrane, redistribute or manufacture what it needs in order to survive, and grow and form new synapses within a more mature, complex environment. While other reviews have focused extensively on the signaling events and cytoskeletal rearrangements that support axon outgrowth and regeneration, we focus this review instead on the underlying membrane trafficking events underlying these processes. Though the mechanisms are still under active investigation, the key roles played by membrane trafficking events during axon repair, growth, and regeneration have been elucidated through elegant comparative studies in both invertebrate and vertebrate organisms. Taken together, a model emerges indicating that the critical requirements for ensuring proper membrane sealing and axon extension include iterative bouts of SNARE mediated exocytosis, endocytosis, and functional links between vesicles and the actin cytoskeleton, similar to the mechanisms utilized during synaptic transmission. This article is part of a Special Issue entitled 'Neuronal Function'.
Collapse
Affiliation(s)
- Ona E Bloom
- The Center for Autoimmune and Musculoskeletal Disease, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY 11030, USA
| | | |
Collapse
|
41
|
Boido D, Farisello P, Cesca F, Ferrea E, Valtorta F, Benfenati F, Baldelli P. Cortico-hippocampal hyperexcitability in synapsin I/II/III knockout mice: age-dependency and response to the antiepileptic drug levetiracetam. Neuroscience 2010; 171:268-83. [PMID: 20804820 DOI: 10.1016/j.neuroscience.2010.08.046] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 08/03/2010] [Accepted: 08/20/2010] [Indexed: 10/19/2022]
Abstract
Synapsins (SynI, SynII, SynIII) are a multigene family of synaptic vesicle (SV) phosphoproteins implicated in the regulation of synaptic transmission and plasticity. Synapsin I, II, I/II and I/II/III knockout mice are epileptic and SYN1/2 genes have been identified as major epilepsy susceptibility genes in humans. We analyzed cortico-hippocampal epileptiform activity induced by 4-aminopyridine (4AP) in acute slices from presymptomatic (3-weeks-old) and symptomatic (1-year-old) Syn I/II/III triple knockout (TKO) mice and aged-matched triple wild type (TWT) controls and assessed the effect of the SV-targeted antiepileptic drug (AED) levetiracetam (LEV) in reverting the epileptic phenotype. Both fast and slow interictal (I-IC) and ictal (IC) events were observed in both genotypes. The incidence of fast I-IC events was higher in presymptomatic TKO slices, while frequency and latency of I-IC events were similar in both genotypes. The major age and genotype effects were observed in IC activity, that was much more pronounced in 3-weeks-old TKO and persisted with age, while it disappeared from 1-year-old TWT slices. LEV virtually suppressed fast I-IC and IC discharges from 3-weeks-old TWT slices, while it only increased the latency of fast I-IC and IC activity in TKO slices. Analysis of I-IC events in patch-clamped CA1 pyramidal neurons revealed that LEV increased the inhibitory/excitatory ratio of I-IC activity in both genotypes. The lower LEV potency in TKO slices of both ages was associated with a decreased expression of SV2A, a SV protein acting as LEV receptor, in cortex and hippocampus. The results demonstrate that deletion of Syn genes is associated with a higher propensity to 4AP-induced epileptic paroxysms that precedes the onset of epilepsy and consolidates with age. LEV ameliorates such hyper excitability by enhancing the inhibition/excitation ratio, although the effect is hindered in TKO slices which exhibit a concomitant decrease in the levels of the LEV receptor SV2A.
Collapse
Affiliation(s)
- D Boido
- Department of Neuroscience and Brain Technologies, The Italian Institute of Technology, Via Morego 30, 16163 Genova, Italy
| | | | | | | | | | | | | |
Collapse
|
42
|
Fornasiero EF, Bonanomi D, Benfenati F, Valtorta F. The role of synapsins in neuronal development. Cell Mol Life Sci 2010; 67:1383-96. [PMID: 20035364 PMCID: PMC11115787 DOI: 10.1007/s00018-009-0227-8] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Revised: 11/22/2009] [Accepted: 12/04/2009] [Indexed: 12/23/2022]
Abstract
The synapsins, the first identified synaptic vesicle-specific proteins, are phosphorylated on multiple sites by a number of protein kinases and are involved in neurite outgrowth and synapse formation as well as in synaptic transmission. In mammals, the synapsin family consists of at least 10 isoforms encoded by 3 distinct genes and composed by a mosaic of conserved and variable domains. The synapsins are highly conserved evolutionarily, and orthologues have been found in invertebrates and lower vertebrates. Within nerve terminals, synapsins are implicated in multiple interactions with presynaptic proteins and the actin cytoskeleton. Via these interactions, synapsins control several mechanisms important for neuronal homeostasis. In this review, we describe the main functional features of the synapsins, in relation to the complex role played by these phosphoproteins in neuronal development.
Collapse
Affiliation(s)
- Eugenio F. Fornasiero
- San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 58, 20132 Milan, Italy
- Unit of Molecular Neuroscience, The Italian Institute of Technology, Via Olgettina 58, 20132 Milan, Italy
| | - Dario Bonanomi
- San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 58, 20132 Milan, Italy
- Present Address: Salk Institute, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
- Unit of Molecular Neuroscience, The Italian Institute of Technology, Via Olgettina 58, 20132 Milan, Italy
| | - Fabio Benfenati
- Department of Neuroscience and Brain Technologies, The Italian Institute of Technology, Via Morego 30, 16163 Genoa, Italy
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genoa, Italy
| | - Flavia Valtorta
- San Raffaele Scientific Institute, Vita-Salute University, Via Olgettina 58, 20132 Milan, Italy
- Unit of Molecular Neuroscience, The Italian Institute of Technology, Via Olgettina 58, 20132 Milan, Italy
| |
Collapse
|
43
|
The highly conserved synapsin domain E mediates synapsin dimerization and phospholipid vesicle clustering. Biochem J 2010; 426:55-64. [PMID: 19922412 DOI: 10.1042/bj20090762] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Synapsins are abundant SV (synaptic vesicle)-associated phosphoproteins that regulate synapse formation and function. The highly conserved C-terminal domain E was shown to contribute to several synapsin functions, ranging from formation of the SV reserve pool to regulation of the kinetics of exocytosis and SV cycling, although the molecular mechanisms underlying these effects are unknown. In the present study, we used a synthetic 25-mer peptide encompassing the most conserved region of domain E (Pep-E) to analyse the role of domain E in regulating the interactions between synapsin I and liposomes mimicking the phospholipid composition of SVs (SV-liposomes) and other pre-synaptic protein partners. In affinity-chromatography and cross-linking assays, Pep-E bound to endogenous and purified exogenous synapsin I and strongly inhibited synapsin dimerization, indicating a role in synapsin oligomerization. Consistently, Pep-E (but not its scrambled version) counteracted the ability of holo-synapsin I to bind and coat phospholipid membranes, as analysed by AFM (atomic force microscopy) topographical scanning, and significantly decreased the clustering of SV-liposomes induced by holo-synapsin I in FRET (Förster resonance energy transfer) assays, suggesting a causal relationship between synapsin oligomerization and vesicle clustering. Either Pep-E or a peptide derived from domain C was necessary and sufficient to inhibit both dimerization and vesicle clustering, indicating the participation of both domains in these activities of synapsin I. The results provide a molecular explanation for the effects of domain E in nerve terminal physiology and suggest that its effects on the size and integrity of SV pools are contributed by the regulation of synapsin dimerization and SV clustering.
Collapse
|
44
|
Porton B, Rodriguiz RM, Phillips LE, Gilbert JW, Feng J, Greengard P, Kao HT, Wetsel WC. Mice lacking synapsin III show abnormalities in explicit memory and conditioned fear. GENES BRAIN AND BEHAVIOR 2009; 9:257-68. [PMID: 20050925 DOI: 10.1111/j.1601-183x.2009.00555.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Synapsin III is a neuron-specific phosphoprotein that plays an important role in synaptic transmission and neural development. While synapsin III is abundant in embryonic brain, expression of the protein in adults is reduced and limited primarily to the hippocampus, olfactory bulb and cerebral cortex. Given the specificity of synapsin III to these brain areas and because it plays a role in neurogenesis in the dentate gyrus, we investigated whether it may affect learning and memory processes in mice. To address this point, synapsin III knockout mice were examined in a general behavioral screen, several tests to assess learning and memory function, and conditioned fear. Mutant animals displayed no anomalies in sensory and motor function or in anxiety- and depressive-like behaviors. Although mutants showed minor alterations in the Morris water maze, they were deficient in object recognition 24 h and 10 days after training and in social transmission of food preference at 20 min and 24 h. In addition, mutants displayed abnormal responses in contextual and cued fear conditioning when tested 1 or 24 h after conditioning. The synapsin III knockout mice also showed aberrant responses in fear-potentiated startle. As synapsin III protein is decreased in schizophrenic brain and because the mutant mice do not harbor obvious anatomical deficits or neurological disorders, these mutants may represent a unique neurodevelopmental model for dissecting the molecular pathways that are related to certain aspects of schizophrenia and related disorders.
Collapse
Affiliation(s)
- B Porton
- Department of Psychiatry and Human Behavior, Brown University, BioMedical Center, Providence, RI 02912, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Role of AP-2alpha transcription factor in the regulation of synapsin II gene expression by dopamine D1 and D2 receptors. J Mol Neurosci 2009; 41:267-77. [PMID: 19842069 DOI: 10.1007/s12031-009-9299-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Accepted: 10/02/2009] [Indexed: 01/11/2023]
Abstract
Synapsins are a family of neuron-specific phosphoproteins involved in synaptic vesicle docking, synaptogenesis, and synaptic plasticity. Previous studies have reported an increase in synapsin II protein by dopaminergic agents in the striatum, medial prefrontal cortex, and nucleus accumbens. This study investigated the mechanistic pathway involved in synapsin II regulation by dopaminergic drugs using primary midbrain neurons to determine which of several transcription factors regulates synapsin II expression. Protein kinase A (PKA) participation in the signaling pathway was examined using selective PKA inhibitors, which reduced synapsin II expression in cell cultures while dopaminergic agents were unable to increase synapsin II in the presence of the PKA inhibitor. Transcription factor involvement was further investigated using separate cultures treated with antisense deoxyoligonucleotides (ADONs) against the following transcription factors: activating protein 2 alpha (AP-2alpha), early growth response factor 1 (EGR-1), or polyoma enhancer activator-3 (PEA-3). Selective knockdown of AP-2alpha by ADONs reduced synapsin II levels, whereas treatment with EGR-1 and PEA-3 ADONs did not affect synapsin II expression. Furthermore, dopaminergic agents were no longer able to influence synapsin II concentrations following AP-2alpha knockdown. Collectively, these results indicate that a cyclic adenosine-3',5'-monophosphate/PKA-dependent mechanism involving the AP-2alpha transcription factor is likely responsible for the increase in neuronal synapsin II following dopamine D1 receptor stimulation or dopamine D2 receptor inhibition.
Collapse
|
46
|
Chen Q, Che R, Wang X, O'Neill FA, Walsh D, Tang W, Shi Y, He L, Kendler KS, Chen X. Association and expression study of synapsin III and schizophrenia. Neurosci Lett 2009; 465:248-51. [PMID: 19766700 DOI: 10.1016/j.neulet.2009.09.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Revised: 08/26/2009] [Accepted: 09/15/2009] [Indexed: 01/27/2023]
Abstract
The synapsin III gene, SYN3, which belongs to the family of synaptic vesicle-associated proteins, has been implicated in the modulation of neurotransmitter release and in synaptogenesis, suggesting a potential role in several neuropsychiatric diseases. The human SYN3 gene is located on chromosome 22q12-13, a candidate region implicated in previous linkage studies of schizophrenia. However, association studies of SYN3 and schizophrenia have produced inconsistent results. In this study, four SYN3 SNPs (rs133945 (-631 C>G), rs133946 (-196 G>A), rs9862 and rs1056484) were tested in three sets of totally 3759 samples that comprise 655 affected subjects and 626 controls in the Irish Case-Control Study of Schizophrenia (ICCSS), 1350 samples incorporating 273 pedigrees in the Irish Study of High Density Schizophrenia Families (ISHDSF), and 564 unrelated schizophrenia patients and 564 healthy individuals in a Chinese case-control sample. The expression levels of SYN3 in schizophrenic patients and unaffected controls were compared using postmortem brain cDNAs provided by the Stanley Medical Research Institute (SMRI). There was no significant association in either the Irish or Chinese case-control samples, nor in the combined samples. Consistent with this finding, we did not find any significant difference in allele or haplotype frequencies when we used the pedigree disequilibrium test to analyze the Irish family sample. In the expression studies, no significant difference (p=0.507) was observed between patients and controls. Both the association studies and expression studies didn't support a major role for SYN3 in the susceptibility of schizophrenia in Irish and Chinese populations.
Collapse
Affiliation(s)
- Qi Chen
- Department of Psychiatry and Virginia Institute for Psychiatric and Behavior Genetics, Virginia Commonwealth University, 800 E. Leigh Street, Richmond, VA 23298, United States.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Transcriptome analysis of nicotine-exposed cells from the brainstem of neonate spontaneously hypertensive and Wistar Kyoto rats. THE PHARMACOGENOMICS JOURNAL 2009; 10:134-60. [DOI: 10.1038/tpj.2009.42] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
48
|
Late-onset dietary restriction compensates for age-related increase in oxidative stress and alterations of HSP 70 and synapsin 1 protein levels in male Wistar rats. Biogerontology 2009; 11:197-209. [PMID: 19609710 DOI: 10.1007/s10522-009-9240-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2009] [Accepted: 06/30/2009] [Indexed: 12/17/2022]
Abstract
Numerous reports implicate increased oxidative stress in the functional and structural changes occurring in the brain and other organs as a part of the normal aging process. Dietary restriction (DR) has long been shown to be life-prolonging intervention in several species. This study was aimed to assess the potential efficacy of late-onset short term DR when initiated in 21 months old male wistar rats for 3 months on the antioxidant defense system and lipid peroxidation, cellular stress response protein HSP 70 and synaptic marker protein synapsin 1 in discrete brain regions such as cortex, hypothalamus, and hippocampus as well as liver, kidney and heart from 24 month old rats. Age-associated decline in activities of superoxide dismutase, catalase, glutathione peroxidase, glutathione, and elevated levels of lipid peroxidation was observed in brain and peripheral organ as well as increased expression of HSP 70 and reduction in synapsin 1 was observed in brain studied. Late-onset short term DR was effective in partially restoring the antioxidant status and in decreasing lipid peroxidation level as well as enhancing the expression of HSP 70 and synapsin 1 in aged rats. Late onset short term DR also prevented age-related neurodegeneration as revealed by Fluoro-Jade B staining in hippocampus and cortex regions of rat brain. Thus our current results suggest that DR initiated even in old age has the potential to improve age related decline in body functions.
Collapse
|
49
|
The importance of synapsin I and II for neurotransmitter levels and vesicular storage in cholinergic, glutamatergic and GABAergic nerve terminals. Neurochem Int 2009; 55:13-21. [DOI: 10.1016/j.neuint.2009.02.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Revised: 02/14/2009] [Accepted: 02/16/2009] [Indexed: 11/20/2022]
|
50
|
Chiappalone M, Casagrande S, Tedesco M, Valtorta F, Baldelli P, Martinoia S, Benfenati F. Opposite Changes in Glutamatergic and GABAergic Transmission Underlie the Diffuse Hyperexcitability of Synapsin I–Deficient Cortical Networks. Cereb Cortex 2008; 19:1422-39. [DOI: 10.1093/cercor/bhn182] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|