1
|
Forte N, Nicois A, Marfella B, Mavaro I, D'Angelo L, Piscitelli F, Scandurra A, De Girolamo P, Baldelli P, Benfenati F, Di Marzo V, Cristino L. Early endocannabinoid-mediated depolarization-induced suppression of excitation delays the appearance of the epileptic phenotype in synapsin II knockout mice. Cell Mol Life Sci 2024; 81:37. [PMID: 38214769 PMCID: PMC11072294 DOI: 10.1007/s00018-023-05029-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/16/2023] [Accepted: 10/31/2023] [Indexed: 01/13/2024]
Abstract
The mechanism underlying the transition from the pre-symptomatic to the symptomatic state is a crucial aspect of epileptogenesis. SYN2 is a member of a multigene family of synaptic vesicle phosphoproteins playing a fundamental role in controlling neurotransmitter release. Human SYN2 gene mutations are associated with epilepsy and autism spectrum disorder. Mice knocked out for synapsin II (SynII KO) are prone to epileptic seizures that appear after 2 months of age. However, the involvement of the endocannabinoid system, known to regulate seizure development and propagation, in the modulation of the excitatory/inhibitory balance in the epileptic hippocampal network of SynII KO mice has not been explored. In this study, we investigated the impact of endocannabinoids on glutamatergic and GABAergic synapses at hippocampal dentate gyrus granule cells in young pre-symptomatic (1-2 months old) and adult symptomatic (5-8 months old) SynII KO mice. We observed an increase in endocannabinoid-mediated depolarization-induced suppression of excitation in young SynII KO mice, compared to age-matched wild-type controls. In contrast, the endocannabinoid-mediated depolarization-induced suppression of inhibition remained unchanged in SynII KO mice at both ages. This selective alteration of excitatory synaptic transmission was accompanied by changes in hippocampal endocannabinoid levels and cannabinoid receptor type 1 distribution among glutamatergic and GABAergic synaptic terminals contacting the granule cells of the dentate gyrus. Finally, inhibition of type-1 cannabinoid receptors in young pre-symptomatic SynII KO mice induced seizures during a tail suspension test. Our results suggest that endocannabinoids contribute to maintaining network stability in a genetic mouse model of human epilepsy.
Collapse
Affiliation(s)
- Nicola Forte
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli (NA), Italy
| | - Alessandro Nicois
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli (NA), Italy
| | - Brenda Marfella
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli (NA), Italy
| | - Isabella Mavaro
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli (NA), Italy
| | - Livia D'Angelo
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Fabiana Piscitelli
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli (NA), Italy
| | - Anna Scandurra
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Paolo De Girolamo
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Pietro Baldelli
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Vincenzo Di Marzo
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli (NA), Italy.
- Faculty of Medicine and Faculty of Agricultural and Food Sciences, Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Québec City, QC, Canada.
- Heart and Lung Research Institute of Université Laval, Québec City, QC, Canada.
- Institute for Nutrition and Functional Foods, Centre NUTRISS, Université Laval, Québec City, QC, Canada.
| | - Luigia Cristino
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli (NA), Italy.
| |
Collapse
|
2
|
Yang Y, Yang J, Liang Y, Zhang G, Cai Z, Zhang Y, Lin H, Tan M. Rab3A interacts with spastin to regulate neurite outgrowth in hippocampal neurons. Biochem Biophys Res Commun 2023; 643:77-87. [PMID: 36587525 DOI: 10.1016/j.bbrc.2022.12.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 12/24/2022]
Abstract
Investigating novel mechanisms of neurite outgrowth via cytoskeleton is critical for developing therapeutic strategies against neural disorders. Rab3A is a vesicle-related protein distributed throughout the nervous system, but the detailed mechanism related to cytoskeleton remains largely unknown. Our previous reports show that spastin serves microtubule to regulate neurite outgrowth. Here, we asked whether Rab3A could function via modulating spastin during neuronal development. The results revealed that Rab3A colocalized with spastin in cultured hippocampal neurons. Immunoprecipitation assays showed that Rab3A physically interacted with spastin in rat brain lysates. Rab3A overexpression significantly induced spastin degradation; this effect was reversed by leupeptin- or MG-132- administration, suggesting the lysosomal and ubiquitin-mediated degradation system. Immunofluorescence staining further confirmed that Rab3A and spastin immune-colocalized with the lysosome marker lysotracker. In COS7 cells, Rab3A overexpression significantly downregulated spastin expression and abolished the spastin-mediated microtubule severing. Furthermore, overexpression inhibited while genetic knockdown of Rab3A promoted neurite outgrowth. However, this inhibitory effect on neurite outgrowth in hippocampal neurons could be reversed via co-transfection of spastin, indicating that Rab3A functions via its interaction protein spastin. In general, our data identify an interaction between Rab3A and spastin, and this interaction affects the protein stability of spastin and eliminates its microtubule severing function, thereby modulating neurite outgrowth.
Collapse
Affiliation(s)
- Yuhao Yang
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Jie Yang
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Yaozhong Liang
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Guowei Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Zhenbin Cai
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Yunlong Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Hongsheng Lin
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China.
| | - Minghui Tan
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China.
| |
Collapse
|
3
|
Michetti C, Falace A, Benfenati F, Fassio A. Synaptic genes and neurodevelopmental disorders: From molecular mechanisms to developmental strategies of behavioral testing. Neurobiol Dis 2022; 173:105856. [PMID: 36070836 DOI: 10.1016/j.nbd.2022.105856] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 10/14/2022] Open
Abstract
Synaptopathies are a class of neurodevelopmental disorders caused by modification in genes coding for synaptic proteins. These proteins oversee the process of neurotransmission, mainly controlling the fusion and recycling of synaptic vesicles at the presynaptic terminal, the expression and localization of receptors at the postsynapse and the coupling between the pre- and the postsynaptic compartments. Murine models, with homozygous or heterozygous deletion for several synaptic genes or knock-in for specific pathogenic mutations, have been developed. They have proved to be extremely informative for understanding synaptic physiology, as well as for clarifying the patho-mechanisms leading to developmental delay, epilepsy and motor, cognitive and social impairments that are the most common clinical manifestations of neurodevelopmental disorders. However, the onset of these disorders emerges during infancy and adolescence while the behavioral phenotyping is often conducted in adult mice, missing important information about the impact of synaptic development and maturation on the manifestation of the behavioral phenotype. Here, we review the main achievements obtained by behavioral testing in murine models of synaptopathies and propose a battery of behavioral tests to improve classification, diagnosis and efficacy of potential therapeutic treatments. Our aim is to underlie the importance of studying behavioral development and better focusing on disease onset and phenotypes.
Collapse
Affiliation(s)
- Caterina Michetti
- Department of Experimental Medicine, University of Genoa, Genoa, Italy; Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia, Genoa, Italy.
| | - Antonio Falace
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Children's Hospital A. Meyer-University of Florence, Florence, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Anna Fassio
- Department of Experimental Medicine, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|
4
|
Schwark R, Andrade R, Bykhovskaia M. Synapsin II Directly Suppresses Epileptic Seizures In Vivo. Brain Sci 2022; 12:brainsci12030325. [PMID: 35326282 PMCID: PMC8946686 DOI: 10.3390/brainsci12030325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 11/21/2022] Open
Abstract
The synapsin family offers a strong linkage between synaptic mechanisms and the epileptic phenotype. Synapsins are phosphoproteins reversibly associated with synaptic vesicles. Synapsin deficiency can cause epilepsy in humans, and synapsin II (SynII) in knockout (KO) mice causes generalized epileptic seizures. To differentiate between the direct effect of SynII versus its secondary adaptations, we used neonatal intracerebroventricular injections of the adeno-associated virus (AAV) expressing SynII. We found that SynII reintroduction diminished the enhanced synaptic activity in Syn2 KO hippocampal slices. Next, we employed the epileptogenic agent 4-aminopyridine (4-AP) and found that SynII reintroduction completely rescued the epileptiform activity observed in Syn2 KO slices upon 4-AP application. Finally, we developed a protocol to provoke behavioral seizures in young Syn2 KO animals and found that SynII reintroduction balances the behavioral seizures. To elucidate the mechanisms through which SynII suppresses hyperexcitability, we injected the phospho-incompetent version of Syn2 that had the mutated protein kinase A (PKA) phosphorylation site. The introduction of the phospho-incompetent SynII mutant suppressed the epileptiform and seizure activity in Syn2 KO mice, but not to the extent observed upon the reintroduction of native SynII. These findings show that SynII can directly suppress seizure activity and that PKA phosphorylation contributes to this function.
Collapse
Affiliation(s)
- Ryan Schwark
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI 48203, USA;
- The Zuckerman Institute, Columbia University, New York, NY 10027, USA
| | - Rodrigo Andrade
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48203, USA;
| | - Maria Bykhovskaia
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI 48203, USA;
- Correspondence:
| |
Collapse
|
5
|
Zimu Z, Jia Z, Xian F, Rui M, Yuting R, Yuan W, Tianhong W, Mian M, Yinlong L, Enfang S. Decreased Expression of PACSIN1 in Brain Glioma Samples Predicts Poor Prognosis. Front Mol Biosci 2021; 8:696072. [PMID: 34422904 PMCID: PMC8375027 DOI: 10.3389/fmolb.2021.696072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/27/2021] [Indexed: 12/03/2022] Open
Abstract
Gliomas are the most severe brain tumours with a poor prognosis. Although surgery, postoperative radiotherapy and chemotherapy can improve the survival rate of glioma patients, the prognosis of most glioma patients is still poor. In recent years, the influence of gene-targeted therapy on gliomas has been gradually discovered, and intervening the occurrence and development of brain gliomas from the perspective of the gene will significantly improve treatment prognosis. Protein Kinase C and Casein Kinase Substrate in Neurons 1 (PACSIN1) is a member of the conserved peripheral membrane protein family in eukaryotes. Improper expression of PACSIN1 can lead to neurological diseases such as Huntington’s disease and schizophrenia. However, its relationship with tumours or even gliomas has not been explored. The study aims to explore PACSIN1 as a prognostic factor that can predict overall survival (OS) for gliomas. We collected the data from CGGA, TCGA, GEO databases and the pathological glioma tissue specimens from 15 clinical glioma patients surgically resected. The differential expression of PACSIN1 in various clinical indicators, the genes related to PACSIN1 expression, the prognostic value of PACSIN1 and the functional annotations and pathway analysis of differently expressed genes (DEGs) were analysed. The results revealed that PACSIN1 had low expression levels in grade IV, IDH1 wild-type and 1p/19q non-codel group gliomas, and PACSIN1 was considered a mesenchymal molecular subtype marker. PACSIN1 expression is positively correlated with OS in all gliomas and it was found that PACSIN1 influenced the occurrence and development of gliomas through synaptic transmission. The PACSIN1 expression is negatively correlated with the malignant degree of gliomas and positively associated with the OS, indicating that PACSIN1 would play an essential role in the occurrence and development of gliomas and might be a potential new biomarker and targeted therapy site for gliomas.
Collapse
Affiliation(s)
- Zhou Zimu
- School of Nursing, Nanjing Medical University, Nanjing, China.,Cancer Nursing Research Branch, Nursing Research Center, Nanjing Medical University, Nanjing, China
| | - Zhang Jia
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Fu Xian
- Department of Neurosurgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Ma Rui
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Ren Yuting
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Wei Yuan
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Wen Tianhong
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Ma Mian
- Department of Neurosurgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Liu Yinlong
- Department of Neurosurgery, The Affiliated Huashan Hospital, Fudan University, Shanghai, China.,Department of Neurosurgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Shan Enfang
- School of Nursing, Nanjing Medical University, Nanjing, China.,Cancer Nursing Research Branch, Nursing Research Center, Nanjing Medical University, Nanjing, China
| |
Collapse
|
6
|
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: 4.0] [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
|
7
|
Zhang Y, Liu Y, Jia Y, Zhao Y, Ma C, Bao X, Meng X, Dou W, Wang X, Ge W. Proteomic profiling of sclerotic hippocampus revealed dysregulated packaging of vesicular neurotransmitters in temporal lobe epilepsy. Epilepsy Res 2020; 166:106412. [DOI: 10.1016/j.eplepsyres.2020.106412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/17/2020] [Accepted: 06/30/2020] [Indexed: 12/31/2022]
|
8
|
Méndez-González MP, Rivera-Aponte DE, Benedikt J, Maldonado-Martínez G, Tejeda-Bayron F, Skatchkov SN, Eaton MJ. Downregulation of Astrocytic Kir4.1 Potassium Channels Is Associated with Hippocampal Neuronal Hyperexcitability in Type 2 Diabetic Mice. Brain Sci 2020; 10:brainsci10020072. [PMID: 32019062 PMCID: PMC7071513 DOI: 10.3390/brainsci10020072] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 01/22/2020] [Indexed: 11/16/2022] Open
Abstract
Epilepsy, characterized by recurrent seizures, affects 1% of the general population. Interestingly, 25% of diabetics develop seizures with a yet unknown mechanism. Hyperglycemia downregulates inwardly rectifying potassium channel 4.1 (Kir4.1) in cultured astrocytes. Therefore, the present study aims to determine if downregulation of functional astrocytic Kir4.1 channels occurs in brains of type 2 diabetic mice and could influence hippocampal neuronal hyperexcitability. Using whole-cell patch clamp recording in hippocampal brain slices from male mice, we determined the electrophysiological properties of stratum radiatum astrocytes and CA1 pyramidal neurons. In diabetic mice, astrocytic Kir4.1 channels were functionally downregulated as evidenced by multiple characteristics including depolarized membrane potential, reduced barium-sensitive Kir currents and impaired potassium uptake capabilities of hippocampal astrocytes. Furthermore, CA1 pyramidal neurons from diabetic mice displayed increased spontaneous activity: action potential frequency was ≈9 times higher in diabetic compared with non-diabetic mice and small EPSC event frequency was significantly higher in CA1 pyramidal cells of diabetics compared to non-diabetics. These differences were apparent in control conditions and largely pronounced in response to the pro-convulsant 4-aminopyridine. Our data suggest that astrocytic dysfunction due to downregulation of Kir4.1 channels may increase seizure susceptibility by impairing astrocytic ability to maintain proper extracellular homeostasis.
Collapse
Affiliation(s)
- Miguel P. Méndez-González
- Department of Biochemistry, Universidad Central del Caribe, Bayamón, PR 00960-6032, USA; (M.P.M.-G.); (F.T.-B.)
- Department of Sciences and Technology, Antilles Adventist University, Mayaguez, PR 00680, USA
- Department of Natural Sciences, University of Puerto Rico, Aguadilla, PR 00604-6150, USA
| | - David E. Rivera-Aponte
- Department of Biochemistry, Universidad Central del Caribe, Bayamón, PR 00960-6032, USA; (M.P.M.-G.); (F.T.-B.)
| | - Jan Benedikt
- Departments of Physiology and Biochemistry Universidad Central del Caribe, Bayamón, PR 00960-6032, USA;
| | | | - Flavia Tejeda-Bayron
- Department of Biochemistry, Universidad Central del Caribe, Bayamón, PR 00960-6032, USA; (M.P.M.-G.); (F.T.-B.)
| | - Serguei N. Skatchkov
- Department of Biochemistry, Universidad Central del Caribe, Bayamón, PR 00960-6032, USA; (M.P.M.-G.); (F.T.-B.)
- Departments of Physiology and Biochemistry Universidad Central del Caribe, Bayamón, PR 00960-6032, USA;
- Correspondence: (S.N.S.); (M.J.E.); Tel.: +787-798-3001 (ext. 2057) (S.N.S.); +787-798-3001 (ext. 2034) (M.J.E.); Fax: +787-786-6285 (M.J.E.)
| | - Misty J. Eaton
- Department of Biochemistry, Universidad Central del Caribe, Bayamón, PR 00960-6032, USA; (M.P.M.-G.); (F.T.-B.)
- Correspondence: (S.N.S.); (M.J.E.); Tel.: +787-798-3001 (ext. 2057) (S.N.S.); +787-798-3001 (ext. 2034) (M.J.E.); Fax: +787-786-6285 (M.J.E.)
| |
Collapse
|
9
|
Matos H, Quiles R, Andrade R, Bykhovskaia M. Growth and excitability at synapsin II deficient hippocampal neurons. Mol Cell Neurosci 2019; 96:25-34. [PMID: 30858140 DOI: 10.1016/j.mcn.2019.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 02/25/2019] [Accepted: 03/07/2019] [Indexed: 10/27/2022] Open
Abstract
Synapsins are neuronal phosphoproteins that fine-tune synaptic transmission and suppress seizure activity. Synapsin II (SynII) deletion produces epileptic seizures and overexcitability in neuronal networks. Early studies in primary neuronal cultures have shown that SynII deletion results in a delay in synapse formation. More recent studies at hippocampal slices have revealed increased spontaneous activity in SynII knockout (SynII(-)) mice. To reconcile these observations, we systematically re-examined synaptic transmission, synapse formation, and neurite growth in primary hippocampal neuronal cultures. We find that spontaneous glutamatergic synaptic activity was suppressed in SynII(-) neurons during the initial developmental epoch (7 days in vitro, DIV) but was enhanced at later times (12 and18 DIV). The density of synapses, transmission between connected pairs of neurons, and the number of docked synaptic vesicles were not affected by SynII deletion. However, we found that neurite outgrowth in SynII(-) neurons was suppressed during the initial developmental epoch (7 DIV) but enhanced at subsequent developmental stages (12 and18 DIV). This finding can account for the observed effect of SynII deletion on synaptic activity. To test whether the observed phenotype resulted directly from the deletion of SynII we expressed SynII in SynII(-) cultures using an adeno-associated virus (AAV). Expression of SynII at 2 DIV rescued the SynII deletion-dependent alterations in both synaptic activity and neuronal growth. To test whether the increased neurite outgrowth in SynII(-) observed at DIV 12 and18 represents an overcompensation for the initial developmental delay or results directly from SynII deletion we performed "late expression" experiments, transfecting SynII(-) cultures with AAV at 7 DIV. The late SynII expression suppressed neurite outgrowth at 12 and 18 DIV to the levels observed in control neurons, suggesting that these phenotypes directly depend on SynII. These results reveal a novel developmentally regulated role for SynII function in the control of neurite growth.
Collapse
Affiliation(s)
- Heidi Matos
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Raymond Quiles
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Rodrigo Andrade
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Maria Bykhovskaia
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI, United States of America.
| |
Collapse
|
10
|
Thomson S, Dyck B, Daya R, Ho J, Bernardo A, Tian Y, Mishra RK. Reduced expression of synapsin II in a chronic phencyclidine preclinical rat model of schizophrenia. Synapse 2018; 73:e22084. [PMID: 30582667 DOI: 10.1002/syn.22084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 12/11/2018] [Accepted: 12/16/2018] [Indexed: 12/25/2022]
Abstract
Schizophrenia is a mental disorder characterized by positive symptoms, negative symptoms, and cognitive dysfunction. Phencyclidine (PCP)-a N-methyl-D-aspartate (NMDA) receptor antagonist-induces symptoms indistinguishable from those of schizophrenia. A reduction of the phosphoprotein synapsin II has also been implicated in schizophrenia and has a well-known role in the maintenance of the presynaptic reserve pool and vesicle mobilization. This study assessed the behavioral and biochemical outcomes of chronic NMDA receptor antagonism in rodents and its implications for the pathophysiology of schizophrenia. Sprague Dawley rats received saline or chronic PCP (5 mg/kg/day) for 14 days via surgically implanted Alzet® osmotic mini-pumps. Following the treatment period, rats were tested with a series of behavioral paradigms, including locomotor activity, social interaction, and sensorimotor gating. Following behavioral assessment, the medial prefrontal cortex (mPFC) of all rats was isolated for synapsin II protein analysis. Chronic PCP treatment yielded a hyper-locomotive state (p = 0.0256), reduced social interaction (p = 0.0005), and reduced pre-pulse inhibition (p < 0.0001) in comparison to saline-treated controls. Synapsin IIa (p < 0.0001) and IIb (p < 0.0071) levels in the mPFC of chronically treated PCP rats were reduced in comparison to the saline group. Study results confirm that rats subject to chronic PCP treatment display behavioral phenotypes similar to established preclinical animal models of schizophrenia. Reduction of synapsin II expression in this context implicates the role of this protein in the pathophysiology of schizophrenia and sheds light on the longer-term consequences of NMDA receptor antagonism facilitated by chronic PCP treatment.
Collapse
Affiliation(s)
- Sharon Thomson
- Faculty of Health Sciences, Department of Psychiatry and Behavioral Neuroscience, McMaster University, Hamilton, Ontario, Canada
| | - Bailey Dyck
- Faculty of Health Sciences, Department of Psychiatry and Behavioral Neuroscience, McMaster University, Hamilton, Ontario, Canada
| | - Ritesh Daya
- Faculty of Health Sciences, Department of Psychiatry and Behavioral Neuroscience, McMaster University, Hamilton, Ontario, Canada
| | - Joella Ho
- Faculty of Health Sciences, Department of Psychiatry and Behavioral Neuroscience, McMaster University, Hamilton, Ontario, Canada
| | - Ashley Bernardo
- Faculty of Health Sciences, Department of Psychiatry and Behavioral Neuroscience, McMaster University, Hamilton, Ontario, Canada
| | - Yuxin Tian
- Faculty of Health Sciences, Department of Psychiatry and Behavioral Neuroscience, McMaster University, Hamilton, Ontario, Canada
| | - Ram K Mishra
- Faculty of Health Sciences, Department of Psychiatry and Behavioral Neuroscience, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
11
|
Ting JT, Lee BR, Chong P, Soler-Llavina G, Cobbs C, Koch C, Zeng H, Lein E. Preparation of Acute Brain Slices Using an Optimized N-Methyl-D-glucamine Protective Recovery Method. J Vis Exp 2018. [PMID: 29553547 DOI: 10.3791/53825] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
This protocol is a practical guide to the N-methyl-D-glucamine (NMDG) protective recovery method of brain slice preparation. Numerous recent studies have validated the utility of this method for enhancing neuronal preservation and overall brain slice viability. The implementation of this technique by early adopters has facilitated detailed investigations into brain function using diverse experimental applications and spanning a wide range of animal ages, brain regions, and cell types. Steps are outlined for carrying out the protective recovery brain slice technique using an optimized NMDG artificial cerebrospinal fluid (aCSF) media formulation and enhanced procedure to reliably obtain healthy brain slices for patch clamp electrophysiology. With this updated approach, a substantial improvement is observed in the speed and reliability of gigaohm seal formation during targeted patch clamp recording experiments while maintaining excellent neuronal preservation, thereby facilitating challenging experimental applications. Representative results are provided from multi-neuron patch clamp recording experiments to assay synaptic connectivity in neocortical brain slices prepared from young adult transgenic mice and mature adult human neurosurgical specimens. Furthermore, the optimized NMDG protective recovery method of brain slicing is compatible with both juvenile and adult animals, thus resolving a limitation of the original methodology. In summary, a single media formulation and brain slicing procedure can be implemented across various species and ages to achieve excellent viability and tissue preservation.
Collapse
Affiliation(s)
| | - Brian R Lee
- Cell Types Program, Allen Institute for Brain Science
| | - Peter Chong
- Cell Types Program, Allen Institute for Brain Science
| | | | - Charles Cobbs
- The Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute
| | - Christof Koch
- Cell Types Program, Allen Institute for Brain Science
| | - Hongkui Zeng
- Cell Types Program, Allen Institute for Brain Science
| | - Ed Lein
- Cell Types Program, Allen Institute for Brain Science
| |
Collapse
|
12
|
Cicvaric A, Bulat T, Bormann D, Yang J, Auer B, Milenkovic I, Cabatic M, Milicevic R, Monje FJ. Sustained consumption of cocoa-based dark chocolate enhances seizure-like events in the mouse hippocampus. Food Funct 2018; 9:1532-1544. [PMID: 29431797 DOI: 10.1039/c7fo01668a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
While the consumption of caffeine and cocoa has been associated with a variety of health benefits to humans, some authors have proposed that excessive caffeine intake may increase the frequency of epileptic seizures in humans and reduce the efficiency of antiepileptic drugs. Little is known, however, about the proconvulsant potential of the sustained, excessive intake of cocoa on hippocampal neural circuits. Using the mouse as an experimental model, we examined the effects of the chronic consumption of food enriched in cocoa-based dark chocolate on motor and mood-related behaviours as well as on the excitability properties of hippocampal neurons. Cocoa food enrichment did not affect body weights or mood-related behaviours but rather promoted general locomotion and improved motor coordination. However, ex vivo electrophysiological analysis revealed a significant enhancement in seizure-like population spike bursting at the neurogenic dentate gyrus, which was paralleled by a significant reduction in the levels of GABA-α1 receptors thus suggesting that an excessive dietary intake of cocoa-enriched food might alter some of the synaptic elements involved in epileptogenesis. These data invite further multidisciplinary research aiming to elucidate the potential deleterious effects of chocolate abuse on behaviour and brain hyperexcitability.
Collapse
Affiliation(s)
- Ana Cicvaric
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| | - Tanja Bulat
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| | - Daniel Bormann
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| | - Jiaye Yang
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| | - Bastian Auer
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| | - Ivan Milenkovic
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Maureen Cabatic
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| | - Radoslav Milicevic
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| | - Francisco J Monje
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| |
Collapse
|
13
|
Altered Development of Synapse Structure and Function in Striatum Caused by Parkinson's Disease-Linked LRRK2-G2019S Mutation. J Neurosci 2017; 36:7128-41. [PMID: 27383589 DOI: 10.1523/jneurosci.3314-15.2016] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 05/26/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Mutations in the gene encoding leucine-rich repeat kinase 2 (LRRK2) can cause Parkinson's disease (PD), and the most common disease-associated mutation, G2019S, increases kinase activity. Because LRRK2 expression levels rise during synaptogenesis and are highest in dorsal striatal spiny projection neurons (SPNs), we tested the hypothesis that the LRRK2-G2019S mutation would alter development of excitatory synaptic networks in dorsal striatum. To circumvent experimental confounds associated with LRRK2 overexpression, we used mice expressing LRRK2-G2019S or D2017A (kinase-dead) knockin mutations. In whole-cell recordings, G2019S SPNs exhibited a fourfold increase in sEPSC frequency compared with wild-type SPNs in postnatal day 21 mice. Such heightened neural activity was increased similarly in direct- and indirect-pathway SPNs, and action potential-dependent activity was particularly elevated. Excitatory synaptic activity in D2017A SPNs was similar to wild type, indicating a selective effect of G2019S. Acute exposure to LRRK2 kinase inhibitors normalized activity, supporting that excessive neural activity in G2019S SPNs is mediated directly and is kinase dependent. Although dendritic arborization and densities of excitatory presynaptic terminals and postsynaptic dendritic spines in G2019S SPNs were similar to wild type, G2019S SPNs displayed larger spines that were matched functionally by a shift toward larger postsynaptic response amplitudes. Acutely isolating striatum from overlying neocortex normalized sEPSC frequency in G2019S mutants, supporting that abnormal corticostriatal activity is involved. These findings indicate that the G2019S mutation imparts a gain-of-abnormal function to SPN activity and morphology during a stage of development when activity can permanently modify circuit structure and function. SIGNIFICANCE STATEMENT Mutations in the kinase domain of leucine-rich repeat kinase 2 (LRRK2) follow Parkinson's disease (PD) heritability. How such mutations affect brain function is poorly understood. LRRK2 expression levels rise after birth at a time when synapses are forming and are highest in dorsal striatum, suggesting that LRRK2 regulates development of striatal circuits. During a period of postnatal development when activity plays a large role in permanently shaping neural circuits, our data show how the most common PD-causing LRRK2 mutation dramatically alters excitatory synaptic activity and the shape of postsynaptic structures in striatum. These findings provide new insight into early functional and structural aberrations in striatal connectivity that may predispose striatal circuitry to both motor and nonmotor dysfunction later in life.
Collapse
|
14
|
Hypertension-induced synapse loss and impairment in synaptic plasticity in the mouse hippocampus mimics the aging phenotype: implications for the pathogenesis of vascular cognitive impairment. GeroScience 2017; 39:385-406. [PMID: 28664509 DOI: 10.1007/s11357-017-9981-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 05/30/2017] [Indexed: 12/22/2022] Open
Abstract
Strong epidemiological and experimental evidence indicates that hypertension has detrimental effects on the cerebral microcirculation and thereby promotes accelerated brain aging. Hypertension is an independent risk factor for both vascular cognitive impairment (VCI) and Alzheimer's disease (AD). However, the pathophysiological link between hypertension-induced cerebromicrovascular injury (e.g., blood-brain barrier disruption, increased microvascular oxidative stress, and inflammation) and cognitive decline remains elusive. The present study was designed to characterize neuronal functional and morphological alterations induced by chronic hypertension and compare them to those induced by aging. To achieve that goal, we induced hypertension in young C57BL/6 mice by chronic (4 weeks) infusion of angiotensin II. We found that long-term potentiation (LTP) of performant path synapses following high-frequency stimulation of afferent fibers was decreased in hippocampal slices obtained from hypertensive mice, mimicking the aging phenotype. Hypertension and advanced age were associated with comparable decline in synaptic density in the stratum radiatum of the mouse hippocampus. Hypertension, similar to aging, was associated with changes in mRNA expression of several genes involved in regulation of neuronal function, including down-regulation of Bdnf, Homer1, and Dlg4, which may have a role in impaired synaptic plasticity. Collectively, hypertension impairs synaptic plasticity, reduces synaptic density, and promotes dysregulation of genes involved in synaptic function in the mouse hippocampus mimicking the aging phenotype. These hypertension-induced neuronal alterations may impair establishment of memories in the hippocampus and contribute to the pathogenesis and clinical manifestation of both vascular cognitive impairment (VCI) and Alzheimer's disease (AD).
Collapse
|
15
|
Li YC, Kavalali ET. Synaptic Vesicle-Recycling Machinery Components as Potential Therapeutic Targets. Pharmacol Rev 2017; 69:141-160. [PMID: 28265000 PMCID: PMC5394918 DOI: 10.1124/pr.116.013342] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Presynaptic nerve terminals are highly specialized vesicle-trafficking machines. Neurotransmitter release from these terminals is sustained by constant local recycling of synaptic vesicles independent from the neuronal cell body. This independence places significant constraints on maintenance of synaptic protein complexes and scaffolds. Key events during the synaptic vesicle cycle-such as exocytosis and endocytosis-require formation and disassembly of protein complexes. This extremely dynamic environment poses unique challenges for proteostasis at synaptic terminals. Therefore, it is not surprising that subtle alterations in synaptic vesicle cycle-associated proteins directly or indirectly contribute to pathophysiology seen in several neurologic and psychiatric diseases. In contrast to the increasing number of examples in which presynaptic dysfunction causes neurologic symptoms or cognitive deficits associated with multiple brain disorders, synaptic vesicle-recycling machinery remains an underexplored drug target. In addition, irrespective of the involvement of presynaptic function in the disease process, presynaptic machinery may also prove to be a viable therapeutic target because subtle alterations in the neurotransmitter release may counter disease mechanisms, correct, or compensate for synaptic communication deficits without the need to interfere with postsynaptic receptor signaling. In this article, we will overview critical properties of presynaptic release machinery to help elucidate novel presynaptic avenues for the development of therapeutic strategies against neurologic and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Ying C Li
- Departments of Neuroscience (Y.C.L., E.T.K.) and Physiology (E.T.K.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ege T Kavalali
- Departments of Neuroscience (Y.C.L., E.T.K.) and Physiology (E.T.K.), University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
16
|
Synapsin II Regulation of GABAergic Synaptic Transmission Is Dependent on Interneuron Subtype. J Neurosci 2017; 37:1757-1771. [PMID: 28087765 DOI: 10.1523/jneurosci.0844-16.2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 12/22/2016] [Accepted: 12/31/2016] [Indexed: 11/21/2022] Open
Abstract
Synapsins are epilepsy susceptibility genes that encode phosphoproteins reversibly associated with synaptic vesicles. Synapsin II (SynII) gene deletion produces a deficit in inhibitory synaptic transmission, and this defect is thought to cause epileptic activity. We systematically investigated how SynII affects synchronous and asynchronous release components of inhibitory transmission in the CA1 region of the mouse hippocampus. We found that the asynchronous GABAergic release component is diminished in SynII-deleted (SynII(-)) slices. To investigate this defect at different interneuron subtypes, we selectively blocked either N-type or P/Q-type Ca2+ channels. SynII deletion suppressed the asynchronous release component at synapses dependent on N-type Ca2+ channels but not at synapses dependent on P/Q-type Ca2+ channels. We then performed paired double-patch recordings from inhibitory basket interneurons connected to pyramidal neurons and used cluster analysis to classify interneurons according to their spiking and synaptic parameters. We identified two cell subtypes, presumably parvalbumin (PV) and cholecystokinin (CCK) expressing basket interneurons. To validate our interneuron classification, we took advantage of transgenic animals with fluorescently labeled PV interneurons and confirmed that their spiking and synaptic parameters matched the parameters of presumed PV cells identified by the cluster analysis. The analysis of the release time course at the two interneuron subtypes demonstrated that the asynchronous release component was selectively reduced at SynII(-) CCK interneurons. In contrast, the transmission was desynchronized at SynII(-) PV interneurons. Together, our results demonstrate that SynII regulates the time course of GABAergic release, and that this SynII function is dependent on the interneuron subtype.SIGNIFICANCE STATEMENT Deletion of the neuronal protein synapsin II (SynII) leads to the development of epilepsy, probably due to impairments in inhibitory synaptic transmission. We systematically investigated SynII function at different subtypes of inhibitory neurons in the hippocampus. We discovered that SynII affects the time course of GABA release, and that this effect is interneuron subtype specific. Within one of the subtypes, SynII deficiency synchronizes the release and suppresses the asynchronous release component, while at the other subtype SynII deficiency suppresses the synchronous release component. These results reveal a new SynII function in the regulation of the time course of GABA release and demonstrate that this function is dependent on the interneuron subtype.
Collapse
|
17
|
Subconvulsant doses of pentylenetetrazol uncover the epileptic phenotype of cultured synapsin-deficient Helix serotonergic neurons in the absence of excitatory and inhibitory inputs. Epilepsy Res 2016; 127:241-251. [PMID: 27639349 DOI: 10.1016/j.eplepsyres.2016.09.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 08/24/2016] [Accepted: 09/06/2016] [Indexed: 11/21/2022]
Abstract
Synapsins are a family of presynaptic proteins related to several processes of synaptic functioning. A variety of reports have linked mutations in synapsin genes with the development of epilepsy. Among the proposed mechanisms, a main one is based on the synapsin-mediated imbalance towards network hyperexcitability due to differential effects on neurotransmitter release in GABAergic and glutamatergic synapses. Along this line, a non-synaptic effect of synapsin depletion increasing neuronal excitability has recently been described in Helix neurons. To further investigate this issue, we examined the effect of synapsin knock-down on the development of pentylenetetrazol (PTZ)-induced epileptic-like activity using single neurons or isolated monosynaptic circuits reconstructed on microelectrode arrays (MEAs). Compared to control neurons, synapsin-silenced neurons showed a lower threshold for the development of epileptic-like activity and prolonged periods of activity, together with the occurrence of spontaneous firing after recurrent PTZ-induced epileptic-like activity. These findings highlight the crucial role of synapsin on neuronal excitability regulation in the absence of inhibitory or excitatory inputs.
Collapse
|
18
|
Kos A, Olde Loohuis N, Meinhardt J, van Bokhoven H, Kaplan BB, Martens GJ, Aschrafi A. MicroRNA-181 promotes synaptogenesis and attenuates axonal outgrowth in cortical neurons. Cell Mol Life Sci 2016; 73:3555-67. [PMID: 27017280 DOI: 10.1007/s00018-016-2179-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 02/13/2016] [Accepted: 03/10/2016] [Indexed: 01/01/2023]
Abstract
MicroRNAs (miRs) are non-coding gene transcripts abundantly expressed in both the developing and adult mammalian brain. They act as important modulators of complex gene regulatory networks during neuronal development and plasticity. miR-181c is highly abundant in cerebellar cortex and its expression is increased in autism patients as well as in an animal model of autism. To systematically identify putative targets of miR-181c, we repressed this miR in growing cortical neurons and found over 70 differentially expressed target genes using transcriptome profiling. Pathway analysis showed that the miR-181c-modulated genes converge on signaling cascades relevant to neurite and synapse developmental processes. To experimentally examine the significance of these data, we inhibited miR-181c during rat cortical neuronal maturation in vitro; this loss-of miR-181c function resulted in enhanced neurite sprouting and reduced synaptogenesis. Collectively, our findings suggest that miR-181c is a modulator of gene networks associated with cortical neuronal maturation.
Collapse
Affiliation(s)
- Aron Kos
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Nikkie Olde Loohuis
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Julia Meinhardt
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Hans van Bokhoven
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Barry B Kaplan
- Laboratory of Molecular Biology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Gerard J Martens
- Department of Molecular Animal Physiology, Radboud University, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Armaz Aschrafi
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands.
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands.
- Laboratory of Molecular Biology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
19
|
Knock-down of synapsin alters cell excitability and action potential waveform by potentiating BK and voltage-gated Ca(2+) currents in Helix serotonergic neurons. Neuroscience 2015; 311:430-43. [PMID: 26522789 DOI: 10.1016/j.neuroscience.2015.10.046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 09/23/2015] [Accepted: 10/25/2015] [Indexed: 11/21/2022]
Abstract
Synapsins (Syns) are an evolutionarily conserved family of presynaptic proteins crucial for the fine-tuning of synaptic function. A large amount of experimental evidences has shown that Syns are involved in the development of epileptic phenotypes and several mutations in Syn genes have been associated with epilepsy in humans and animal models. Syn mutations induce alterations in circuitry and neurotransmitter release, differentially affecting excitatory and inhibitory synapses, thus causing an excitation/inhibition imbalance in network excitability toward hyperexcitability that may be a determinant with regard to the development of epilepsy. Another approach to investigate epileptogenic mechanisms is to understand how silencing Syn affects the cellular behavior of single neurons and is associated with the hyperexcitable phenotypes observed in epilepsy. Here, we examined the functional effects of antisense-RNA inhibition of Syn expression on individually identified and isolated serotonergic cells of the Helix land snail. We found that Helix synapsin silencing increases cell excitability characterized by a slightly depolarized resting membrane potential, decreases the rheobase, reduces the threshold for action potential (AP) firing and increases the mean and instantaneous firing rates, with respect to control cells. The observed increase of Ca(2+) and BK currents in Syn-silenced cells seems to be related to changes in the shape of the AP waveform. These currents sustain the faster spiking in Syn-deficient cells by increasing the after hyperpolarization and limiting the Na(+) and Ca(2+) channel inactivation during repetitive firing. This in turn speeds up the depolarization phase by reaching the AP threshold faster. Our results provide evidence that Syn silencing increases intrinsic cell excitability associated with increased Ca(2+) and Ca(2+)-dependent BK currents in the absence of excitatory or inhibitory inputs.
Collapse
|
20
|
Moreira-Filho CA, Bando SY, Bertonha FB, Iamashita P, Silva FN, Costa LDF, Silva AV, Castro LHM, Wen HT. Community structure analysis of transcriptional networks reveals distinct molecular pathways for early- and late-onset temporal lobe epilepsy with childhood febrile seizures. PLoS One 2015; 10:e0128174. [PMID: 26011637 PMCID: PMC4444281 DOI: 10.1371/journal.pone.0128174] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 04/24/2015] [Indexed: 12/21/2022] Open
Abstract
Age at epilepsy onset has a broad impact on brain plasticity and epilepsy pathomechanisms. Prolonged febrile seizures in early childhood (FS) constitute an initial precipitating insult (IPI) commonly associated with mesial temporal lobe epilepsy (MTLE). FS-MTLE patients may have early disease onset, i.e. just after the IPI, in early childhood, or late-onset, ranging from mid-adolescence to early adult life. The mechanisms governing early (E) or late (L) disease onset are largely unknown. In order to unveil the molecular pathways underlying E and L subtypes of FS-MTLE we investigated global gene expression in hippocampal CA3 explants of FS-MTLE patients submitted to hippocampectomy. Gene coexpression networks (GCNs) were obtained for the E and L patient groups. A network-based approach for GCN analysis was employed allowing: i) the visualization and analysis of differentially expressed (DE) and complete (CO) - all valid GO annotated transcripts - GCNs for the E and L groups; ii) the study of interactions between all the system's constituents based on community detection and coarse-grained community structure methods. We found that the E-DE communities with strongest connection weights harbor highly connected genes mainly related to neural excitability and febrile seizures, whereas in L-DE communities these genes are not only involved in network excitability but also playing roles in other epilepsy-related processes. Inversely, in E-CO the strongly connected communities are related to compensatory pathways (seizure inhibition, neuronal survival and responses to stress conditions) while in L-CO these communities harbor several genes related to pro-epileptic effects, seizure-related mechanisms and vulnerability to epilepsy. These results fit the concept, based on fMRI and behavioral studies, that early onset epilepsies, although impacting more severely the hippocampus, are associated to compensatory mechanisms, while in late MTLE development the brain is less able to generate adaptive mechanisms, what has implications for epilepsy management and drug discovery.
Collapse
Affiliation(s)
| | - Silvia Yumi Bando
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Fernanda Bernardi Bertonha
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Priscila Iamashita
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | | | | | | | - Luiz Henrique Martins Castro
- Department of Neurology, FMUSP, São Paulo, SP, Brazil
- Clinical Neurology Division, Hospital das Clínicas, FMUSP, São Paulo, SP, Brazil
| | - Hung-Tzu Wen
- Epilepsy Surgery Group, Hospital das Clínicas, FMUSP, São Paulo, SP, Brazil
| |
Collapse
|