Cargando…

Long non-coding RNA ANRIL knockdown suppresses apoptosis and pro-inflammatory cytokines while enhancing neurite outgrowth via binding microRNA-125a in a cellular model of Alzheimer's disease

The present study aimed to investigate the effect of the long non-coding RNA antisense non-coding RNA in the INK4 locus (lnc-ANRIL) knockdown on apoptosis, neurite outgrowth and inflammation based on a PC12 cellular Alzheimer's disease (AD) model. A cellular AD model was constructed by treating...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhou, Bingling, Li, Lijuan, Qiu, Xin, Wu, Jiashun, Xu, Lei, Shao, Wei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7339647/
https://www.ncbi.nlm.nih.gov/pubmed/32626959
http://dx.doi.org/10.3892/mmr.2020.11203
_version_ 1783554928739876864
author Zhou, Bingling
Li, Lijuan
Qiu, Xin
Wu, Jiashun
Xu, Lei
Shao, Wei
author_facet Zhou, Bingling
Li, Lijuan
Qiu, Xin
Wu, Jiashun
Xu, Lei
Shao, Wei
author_sort Zhou, Bingling
collection PubMed
description The present study aimed to investigate the effect of the long non-coding RNA antisense non-coding RNA in the INK4 locus (lnc-ANRIL) knockdown on apoptosis, neurite outgrowth and inflammation based on a PC12 cellular Alzheimer's disease (AD) model. A cellular AD model was constructed by treating nerve growth factor stimulated PC12 cells with amyloid β (Aβ) 1–42 and then control knockdown plasmid and lnc-ANRIL knockdown plasmid were transfected in the PC12 cellular AD model as the KD- negative control (NC) group or the AD-ANRIL group respectively. Apoptosis, neurite outgrowth, pro-inflammatory cytokines and microRNA (miR)-125a were assessed. Rescue experiments were conducted by transfecting lnc-ANRIL knockdown plasmid and lnc-ANRIL knockdown plasmid and miR-125a inhibitor in the PC12 cellular AD model as the KD-ANRIL group or KD-ANRIL + KD-miR-125a group respectively. Following transfection, cell apoptosis deccreased while neurite outgrowth increased in the KD-ANRIL group compared with the KD-NC group (all P<0.01). Concerning inflammation, tumor necrosis factor-α (TNF-α) and interleukin (IL)-1β, IL-6 and IL-17 were decreased in the KD-ANRIL group compared with the KD-NC group (all P<0.01). miR-125a was negatively regulated by lnc-ANRIL and therefore rescue experiments were subsequently conducted. In the rescue experiments, cell apoptosis was increased while total neurite outgrowth was inhibited in the KD-ANRIL + KD-miR-125a group compared with the KD-ANRIL group (all P<0.01), and TNF-α, IL-1β, IL-6 and IL-17 were increased in the KD-ANRIL + KD-miR-125a group compared with the KD-ANRIL group (all P<0.01). A luciferase reporter assay demonstrated that lnc-ANRIL directly bound miR-125a. lnc-ANRIL knockdown suppressed cell apoptosis and inflammation while promoting neurite outgrowth via binding of miR-125a in AD.
format Online
Article
Text
id pubmed-7339647
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-73396472020-07-09 Long non-coding RNA ANRIL knockdown suppresses apoptosis and pro-inflammatory cytokines while enhancing neurite outgrowth via binding microRNA-125a in a cellular model of Alzheimer's disease Zhou, Bingling Li, Lijuan Qiu, Xin Wu, Jiashun Xu, Lei Shao, Wei Mol Med Rep Articles The present study aimed to investigate the effect of the long non-coding RNA antisense non-coding RNA in the INK4 locus (lnc-ANRIL) knockdown on apoptosis, neurite outgrowth and inflammation based on a PC12 cellular Alzheimer's disease (AD) model. A cellular AD model was constructed by treating nerve growth factor stimulated PC12 cells with amyloid β (Aβ) 1–42 and then control knockdown plasmid and lnc-ANRIL knockdown plasmid were transfected in the PC12 cellular AD model as the KD- negative control (NC) group or the AD-ANRIL group respectively. Apoptosis, neurite outgrowth, pro-inflammatory cytokines and microRNA (miR)-125a were assessed. Rescue experiments were conducted by transfecting lnc-ANRIL knockdown plasmid and lnc-ANRIL knockdown plasmid and miR-125a inhibitor in the PC12 cellular AD model as the KD-ANRIL group or KD-ANRIL + KD-miR-125a group respectively. Following transfection, cell apoptosis deccreased while neurite outgrowth increased in the KD-ANRIL group compared with the KD-NC group (all P<0.01). Concerning inflammation, tumor necrosis factor-α (TNF-α) and interleukin (IL)-1β, IL-6 and IL-17 were decreased in the KD-ANRIL group compared with the KD-NC group (all P<0.01). miR-125a was negatively regulated by lnc-ANRIL and therefore rescue experiments were subsequently conducted. In the rescue experiments, cell apoptosis was increased while total neurite outgrowth was inhibited in the KD-ANRIL + KD-miR-125a group compared with the KD-ANRIL group (all P<0.01), and TNF-α, IL-1β, IL-6 and IL-17 were increased in the KD-ANRIL + KD-miR-125a group compared with the KD-ANRIL group (all P<0.01). A luciferase reporter assay demonstrated that lnc-ANRIL directly bound miR-125a. lnc-ANRIL knockdown suppressed cell apoptosis and inflammation while promoting neurite outgrowth via binding of miR-125a in AD. D.A. Spandidos 2020-08 2020-06-02 /pmc/articles/PMC7339647/ /pubmed/32626959 http://dx.doi.org/10.3892/mmr.2020.11203 Text en Copyright: © Zhou et al. This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License (https://creativecommons.org/licenses/by-nc-nd/4.0/) , which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
spellingShingle Articles
Zhou, Bingling
Li, Lijuan
Qiu, Xin
Wu, Jiashun
Xu, Lei
Shao, Wei
Long non-coding RNA ANRIL knockdown suppresses apoptosis and pro-inflammatory cytokines while enhancing neurite outgrowth via binding microRNA-125a in a cellular model of Alzheimer's disease
title Long non-coding RNA ANRIL knockdown suppresses apoptosis and pro-inflammatory cytokines while enhancing neurite outgrowth via binding microRNA-125a in a cellular model of Alzheimer's disease
title_full Long non-coding RNA ANRIL knockdown suppresses apoptosis and pro-inflammatory cytokines while enhancing neurite outgrowth via binding microRNA-125a in a cellular model of Alzheimer's disease
title_fullStr Long non-coding RNA ANRIL knockdown suppresses apoptosis and pro-inflammatory cytokines while enhancing neurite outgrowth via binding microRNA-125a in a cellular model of Alzheimer's disease
title_full_unstemmed Long non-coding RNA ANRIL knockdown suppresses apoptosis and pro-inflammatory cytokines while enhancing neurite outgrowth via binding microRNA-125a in a cellular model of Alzheimer's disease
title_short Long non-coding RNA ANRIL knockdown suppresses apoptosis and pro-inflammatory cytokines while enhancing neurite outgrowth via binding microRNA-125a in a cellular model of Alzheimer's disease
title_sort long non-coding rna anril knockdown suppresses apoptosis and pro-inflammatory cytokines while enhancing neurite outgrowth via binding microrna-125a in a cellular model of alzheimer's disease
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7339647/
https://www.ncbi.nlm.nih.gov/pubmed/32626959
http://dx.doi.org/10.3892/mmr.2020.11203
work_keys_str_mv AT zhoubingling longnoncodingrnaanrilknockdownsuppressesapoptosisandproinflammatorycytokineswhileenhancingneuriteoutgrowthviabindingmicrorna125ainacellularmodelofalzheimersdisease
AT lilijuan longnoncodingrnaanrilknockdownsuppressesapoptosisandproinflammatorycytokineswhileenhancingneuriteoutgrowthviabindingmicrorna125ainacellularmodelofalzheimersdisease
AT qiuxin longnoncodingrnaanrilknockdownsuppressesapoptosisandproinflammatorycytokineswhileenhancingneuriteoutgrowthviabindingmicrorna125ainacellularmodelofalzheimersdisease
AT wujiashun longnoncodingrnaanrilknockdownsuppressesapoptosisandproinflammatorycytokineswhileenhancingneuriteoutgrowthviabindingmicrorna125ainacellularmodelofalzheimersdisease
AT xulei longnoncodingrnaanrilknockdownsuppressesapoptosisandproinflammatorycytokineswhileenhancingneuriteoutgrowthviabindingmicrorna125ainacellularmodelofalzheimersdisease
AT shaowei longnoncodingrnaanrilknockdownsuppressesapoptosisandproinflammatorycytokineswhileenhancingneuriteoutgrowthviabindingmicrorna125ainacellularmodelofalzheimersdisease