Cargando…

Recombinant CCL17-dependent CCR4 activation alleviates neuroinflammation and neuronal apoptosis through the PI3K/AKT/Foxo1 signaling pathway after ICH in mice

BACKGROUND: Intracerebral hemorrhage (ICH), a devastating subtype of stroke, is associated with high mortality and morbidity. Neuroinflammation is an important factor leading to ICH-induced neurological injuries. C-C Chemokine Receptor 4 (CCR4) plays an important role in enhancing hematoma clearance...

Descripción completa

Detalles Bibliográficos
Autores principales: Deng, Shuixiang, Jin, Peng, Sherchan, Prativa, Liu, Shengpeng, Cui, Yuhui, Huang, Lei, Zhang, John H., Gong, Ye, Tang, Jiping
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7923481/
https://www.ncbi.nlm.nih.gov/pubmed/33648537
http://dx.doi.org/10.1186/s12974-021-02112-3
_version_ 1783658911435325440
author Deng, Shuixiang
Jin, Peng
Sherchan, Prativa
Liu, Shengpeng
Cui, Yuhui
Huang, Lei
Zhang, John H.
Gong, Ye
Tang, Jiping
author_facet Deng, Shuixiang
Jin, Peng
Sherchan, Prativa
Liu, Shengpeng
Cui, Yuhui
Huang, Lei
Zhang, John H.
Gong, Ye
Tang, Jiping
author_sort Deng, Shuixiang
collection PubMed
description BACKGROUND: Intracerebral hemorrhage (ICH), a devastating subtype of stroke, is associated with high mortality and morbidity. Neuroinflammation is an important factor leading to ICH-induced neurological injuries. C-C Chemokine Receptor 4 (CCR4) plays an important role in enhancing hematoma clearance after ICH. However, it is unclear whether CCR4 activation can ameliorate neuroinflammation and apoptosis of neurons following ICH. The aim of the present study was to examine the effects of recombinant CCL17 (rCCL17)-dependent CCR4 activation on neuroinflammation and neuronal apoptosis in an intrastriatal autologous blood injection ICH model, and to determine whether the PI3K/AKT/Foxo1 signaling pathway was involved. METHODS: Two hundred twenty-six adult (8-week-old) male CD1 mice were randomly assigned to sham and ICH surgery groups. An intrastriatal autologous blood injection ICH model was used. rCCL17, a CCR4 ligand, was delivered by intranasal administration at 1 h, 3 h, and 6 h post-ICH. CCL17 antibody was administrated by intraventricular injection at 1 h post-ICH. C021, a specific inhibitor of CCR4 and GDC0068, an AKT inhibitor were delivered intraperitoneally 1 h prior to ICH induction. Brain edema, neurobehavioral assessments, western blotting, Fluoro-Jade C staining, terminal deoxynucleotidyl transferase dUTP nick end labeling, and immunofluorescence staining were conducted. RESULTS: Endogenous expression of CCL17 and CCR4 were increased following ICH, peaking at 5 days post-induction. CCR4 was found to co-localize with microglia, neurons, and astrocytes. rCCL17 treatment decreased brain water content, attenuated short- and long-term neurological deficits, deceased activation of microglia/macrophages and infiltration of neutrophils, and inhibited neuronal apoptosis in the perihematomal region post-ICH. Moreover, rCCL17 treatment post-ICH significantly increased the expression of CCR4, PI3K, phosphorylated AKT, and Bcl-2, while Foxo1, IL-1β, TNF-α, and Bax expression were decreased. The neuroprotective effects of rCCL17 were reversed with the administration of C021 or GDC0068. CONCLUSIONS: rCCL17-dependent CCR4 activation ameliorated neurological deficits, reduced brain edema, and ameliorated neuroinflammation and neuronal apoptosis, at least in part, through the PI3K/AKT/Foxo1 signaling pathway after ICH. Thus, activation of CCR4 may provide a promising therapeutic approach for the early management of ICH. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12974-021-02112-3.
format Online
Article
Text
id pubmed-7923481
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-79234812021-03-02 Recombinant CCL17-dependent CCR4 activation alleviates neuroinflammation and neuronal apoptosis through the PI3K/AKT/Foxo1 signaling pathway after ICH in mice Deng, Shuixiang Jin, Peng Sherchan, Prativa Liu, Shengpeng Cui, Yuhui Huang, Lei Zhang, John H. Gong, Ye Tang, Jiping J Neuroinflammation Research BACKGROUND: Intracerebral hemorrhage (ICH), a devastating subtype of stroke, is associated with high mortality and morbidity. Neuroinflammation is an important factor leading to ICH-induced neurological injuries. C-C Chemokine Receptor 4 (CCR4) plays an important role in enhancing hematoma clearance after ICH. However, it is unclear whether CCR4 activation can ameliorate neuroinflammation and apoptosis of neurons following ICH. The aim of the present study was to examine the effects of recombinant CCL17 (rCCL17)-dependent CCR4 activation on neuroinflammation and neuronal apoptosis in an intrastriatal autologous blood injection ICH model, and to determine whether the PI3K/AKT/Foxo1 signaling pathway was involved. METHODS: Two hundred twenty-six adult (8-week-old) male CD1 mice were randomly assigned to sham and ICH surgery groups. An intrastriatal autologous blood injection ICH model was used. rCCL17, a CCR4 ligand, was delivered by intranasal administration at 1 h, 3 h, and 6 h post-ICH. CCL17 antibody was administrated by intraventricular injection at 1 h post-ICH. C021, a specific inhibitor of CCR4 and GDC0068, an AKT inhibitor were delivered intraperitoneally 1 h prior to ICH induction. Brain edema, neurobehavioral assessments, western blotting, Fluoro-Jade C staining, terminal deoxynucleotidyl transferase dUTP nick end labeling, and immunofluorescence staining were conducted. RESULTS: Endogenous expression of CCL17 and CCR4 were increased following ICH, peaking at 5 days post-induction. CCR4 was found to co-localize with microglia, neurons, and astrocytes. rCCL17 treatment decreased brain water content, attenuated short- and long-term neurological deficits, deceased activation of microglia/macrophages and infiltration of neutrophils, and inhibited neuronal apoptosis in the perihematomal region post-ICH. Moreover, rCCL17 treatment post-ICH significantly increased the expression of CCR4, PI3K, phosphorylated AKT, and Bcl-2, while Foxo1, IL-1β, TNF-α, and Bax expression were decreased. The neuroprotective effects of rCCL17 were reversed with the administration of C021 or GDC0068. CONCLUSIONS: rCCL17-dependent CCR4 activation ameliorated neurological deficits, reduced brain edema, and ameliorated neuroinflammation and neuronal apoptosis, at least in part, through the PI3K/AKT/Foxo1 signaling pathway after ICH. Thus, activation of CCR4 may provide a promising therapeutic approach for the early management of ICH. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12974-021-02112-3. BioMed Central 2021-03-01 /pmc/articles/PMC7923481/ /pubmed/33648537 http://dx.doi.org/10.1186/s12974-021-02112-3 Text en © The Author(s) 2021 Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Deng, Shuixiang
Jin, Peng
Sherchan, Prativa
Liu, Shengpeng
Cui, Yuhui
Huang, Lei
Zhang, John H.
Gong, Ye
Tang, Jiping
Recombinant CCL17-dependent CCR4 activation alleviates neuroinflammation and neuronal apoptosis through the PI3K/AKT/Foxo1 signaling pathway after ICH in mice
title Recombinant CCL17-dependent CCR4 activation alleviates neuroinflammation and neuronal apoptosis through the PI3K/AKT/Foxo1 signaling pathway after ICH in mice
title_full Recombinant CCL17-dependent CCR4 activation alleviates neuroinflammation and neuronal apoptosis through the PI3K/AKT/Foxo1 signaling pathway after ICH in mice
title_fullStr Recombinant CCL17-dependent CCR4 activation alleviates neuroinflammation and neuronal apoptosis through the PI3K/AKT/Foxo1 signaling pathway after ICH in mice
title_full_unstemmed Recombinant CCL17-dependent CCR4 activation alleviates neuroinflammation and neuronal apoptosis through the PI3K/AKT/Foxo1 signaling pathway after ICH in mice
title_short Recombinant CCL17-dependent CCR4 activation alleviates neuroinflammation and neuronal apoptosis through the PI3K/AKT/Foxo1 signaling pathway after ICH in mice
title_sort recombinant ccl17-dependent ccr4 activation alleviates neuroinflammation and neuronal apoptosis through the pi3k/akt/foxo1 signaling pathway after ich in mice
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7923481/
https://www.ncbi.nlm.nih.gov/pubmed/33648537
http://dx.doi.org/10.1186/s12974-021-02112-3
work_keys_str_mv AT dengshuixiang recombinantccl17dependentccr4activationalleviatesneuroinflammationandneuronalapoptosisthroughthepi3kaktfoxo1signalingpathwayafterichinmice
AT jinpeng recombinantccl17dependentccr4activationalleviatesneuroinflammationandneuronalapoptosisthroughthepi3kaktfoxo1signalingpathwayafterichinmice
AT sherchanprativa recombinantccl17dependentccr4activationalleviatesneuroinflammationandneuronalapoptosisthroughthepi3kaktfoxo1signalingpathwayafterichinmice
AT liushengpeng recombinantccl17dependentccr4activationalleviatesneuroinflammationandneuronalapoptosisthroughthepi3kaktfoxo1signalingpathwayafterichinmice
AT cuiyuhui recombinantccl17dependentccr4activationalleviatesneuroinflammationandneuronalapoptosisthroughthepi3kaktfoxo1signalingpathwayafterichinmice
AT huanglei recombinantccl17dependentccr4activationalleviatesneuroinflammationandneuronalapoptosisthroughthepi3kaktfoxo1signalingpathwayafterichinmice
AT zhangjohnh recombinantccl17dependentccr4activationalleviatesneuroinflammationandneuronalapoptosisthroughthepi3kaktfoxo1signalingpathwayafterichinmice
AT gongye recombinantccl17dependentccr4activationalleviatesneuroinflammationandneuronalapoptosisthroughthepi3kaktfoxo1signalingpathwayafterichinmice
AT tangjiping recombinantccl17dependentccr4activationalleviatesneuroinflammationandneuronalapoptosisthroughthepi3kaktfoxo1signalingpathwayafterichinmice