Cargando…

Silencing miR‐20a‐5p inhibits axonal growth and neuronal branching and prevents epileptogenesis through RGMa‐RhoA‐mediated synaptic plasticity

Epileptogenesis is a potential process. Mossy fibre sprouting (MFS) and synaptic plasticity promote epileptogenesis. Overexpression of repulsive guidance molecule a (RGMa) prevents epileptogenesis by inhibiting MFS. However, other aspects underlying the RGMa regulatory process of epileptogenesis hav...

Descripción completa

Detalles Bibliográficos
Autores principales: Feng, Yanyan, Duan, Chaojun, Luo, Zhaohui, Xiao, Wenbiao, Tian, Fafa
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7521253/
https://www.ncbi.nlm.nih.gov/pubmed/32779334
http://dx.doi.org/10.1111/jcmm.15677
_version_ 1783587939088859136
author Feng, Yanyan
Duan, Chaojun
Luo, Zhaohui
Xiao, Wenbiao
Tian, Fafa
author_facet Feng, Yanyan
Duan, Chaojun
Luo, Zhaohui
Xiao, Wenbiao
Tian, Fafa
author_sort Feng, Yanyan
collection PubMed
description Epileptogenesis is a potential process. Mossy fibre sprouting (MFS) and synaptic plasticity promote epileptogenesis. Overexpression of repulsive guidance molecule a (RGMa) prevents epileptogenesis by inhibiting MFS. However, other aspects underlying the RGMa regulatory process of epileptogenesis have not been elucidated. We studied whether RGMa could be modulated by microRNAs and regulated RhoA in epileptogenesis. Using microRNA databases, we selected four miRNAs as potential candidates. We further experimentally confirmed miR‐20a‐5p as a RGMa upstream regulator. Then, in vitro, by manipulating miR‐20a‐5p and RGMa, we investigated the regulatory relationship between miR‐20a‐5p, RGMa and RhoA, and the effects of this pathway on neuronal morphology. Finally, in the epilepsy animal model, we determined whether the miR‐20a‐5p‐RGMa‐RhoA pathway influenced MFS and synaptic plasticity and then modified epileptogenesis. Our results showed that miR‐20a‐5p regulated RGMa and that RGMa regulated RhoA in vitro. Furthermore, in primary hippocampal neurons, the miR‐20a‐5p‐RGMa‐RhoA pathway regulated axonal growth and neuronal branching; in the PTZ‐induced epilepsy model, silencing miR‐20a‐5p prevented epileptogenesis through RGMa‐RhoA‐mediated synaptic plasticity but did not change MFS. Overall, we concluded that silencing miR‐20a‐5p inhibits axonal growth and neuronal branching and prevents epileptogenesis through RGMa‐RhoA‐mediated synaptic plasticity in the PTZ‐induced epilepsy model, thereby providing a possible strategy to prevent epileptogenesis.
format Online
Article
Text
id pubmed-7521253
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-75212532020-09-30 Silencing miR‐20a‐5p inhibits axonal growth and neuronal branching and prevents epileptogenesis through RGMa‐RhoA‐mediated synaptic plasticity Feng, Yanyan Duan, Chaojun Luo, Zhaohui Xiao, Wenbiao Tian, Fafa J Cell Mol Med Original Articles Epileptogenesis is a potential process. Mossy fibre sprouting (MFS) and synaptic plasticity promote epileptogenesis. Overexpression of repulsive guidance molecule a (RGMa) prevents epileptogenesis by inhibiting MFS. However, other aspects underlying the RGMa regulatory process of epileptogenesis have not been elucidated. We studied whether RGMa could be modulated by microRNAs and regulated RhoA in epileptogenesis. Using microRNA databases, we selected four miRNAs as potential candidates. We further experimentally confirmed miR‐20a‐5p as a RGMa upstream regulator. Then, in vitro, by manipulating miR‐20a‐5p and RGMa, we investigated the regulatory relationship between miR‐20a‐5p, RGMa and RhoA, and the effects of this pathway on neuronal morphology. Finally, in the epilepsy animal model, we determined whether the miR‐20a‐5p‐RGMa‐RhoA pathway influenced MFS and synaptic plasticity and then modified epileptogenesis. Our results showed that miR‐20a‐5p regulated RGMa and that RGMa regulated RhoA in vitro. Furthermore, in primary hippocampal neurons, the miR‐20a‐5p‐RGMa‐RhoA pathway regulated axonal growth and neuronal branching; in the PTZ‐induced epilepsy model, silencing miR‐20a‐5p prevented epileptogenesis through RGMa‐RhoA‐mediated synaptic plasticity but did not change MFS. Overall, we concluded that silencing miR‐20a‐5p inhibits axonal growth and neuronal branching and prevents epileptogenesis through RGMa‐RhoA‐mediated synaptic plasticity in the PTZ‐induced epilepsy model, thereby providing a possible strategy to prevent epileptogenesis. John Wiley and Sons Inc. 2020-08-10 2020-09 /pmc/articles/PMC7521253/ /pubmed/32779334 http://dx.doi.org/10.1111/jcmm.15677 Text en © 2020 The Authors. Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Original Articles
Feng, Yanyan
Duan, Chaojun
Luo, Zhaohui
Xiao, Wenbiao
Tian, Fafa
Silencing miR‐20a‐5p inhibits axonal growth and neuronal branching and prevents epileptogenesis through RGMa‐RhoA‐mediated synaptic plasticity
title Silencing miR‐20a‐5p inhibits axonal growth and neuronal branching and prevents epileptogenesis through RGMa‐RhoA‐mediated synaptic plasticity
title_full Silencing miR‐20a‐5p inhibits axonal growth and neuronal branching and prevents epileptogenesis through RGMa‐RhoA‐mediated synaptic plasticity
title_fullStr Silencing miR‐20a‐5p inhibits axonal growth and neuronal branching and prevents epileptogenesis through RGMa‐RhoA‐mediated synaptic plasticity
title_full_unstemmed Silencing miR‐20a‐5p inhibits axonal growth and neuronal branching and prevents epileptogenesis through RGMa‐RhoA‐mediated synaptic plasticity
title_short Silencing miR‐20a‐5p inhibits axonal growth and neuronal branching and prevents epileptogenesis through RGMa‐RhoA‐mediated synaptic plasticity
title_sort silencing mir‐20a‐5p inhibits axonal growth and neuronal branching and prevents epileptogenesis through rgma‐rhoa‐mediated synaptic plasticity
topic Original Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7521253/
https://www.ncbi.nlm.nih.gov/pubmed/32779334
http://dx.doi.org/10.1111/jcmm.15677
work_keys_str_mv AT fengyanyan silencingmir20a5pinhibitsaxonalgrowthandneuronalbranchingandpreventsepileptogenesisthroughrgmarhoamediatedsynapticplasticity
AT duanchaojun silencingmir20a5pinhibitsaxonalgrowthandneuronalbranchingandpreventsepileptogenesisthroughrgmarhoamediatedsynapticplasticity
AT luozhaohui silencingmir20a5pinhibitsaxonalgrowthandneuronalbranchingandpreventsepileptogenesisthroughrgmarhoamediatedsynapticplasticity
AT xiaowenbiao silencingmir20a5pinhibitsaxonalgrowthandneuronalbranchingandpreventsepileptogenesisthroughrgmarhoamediatedsynapticplasticity
AT tianfafa silencingmir20a5pinhibitsaxonalgrowthandneuronalbranchingandpreventsepileptogenesisthroughrgmarhoamediatedsynapticplasticity