Cargando…

Apremilast Regulates the Teff/Treg Balance to Ameliorate Uveitis via PI3K/AKT/FoxO1 Signaling Pathway

Autoimmune uveitis (AU), being one of the sight-threatening ocular inflammatory disorders, has been widely regarded by ophthalmologists and immunologists as a great challenge. Apremilast, a phosphodiesterase-4 inhibitor (PDE4i), which was approved by the U.S. Food and Drug Administration (FDA) for t...

Descripción completa

Detalles Bibliográficos
Autores principales: Chen, Yuxi, Li, Zhuang, Li, He, Su, Wenru, Xie, Yanyan, Pan, Yuan, Chen, Xiaoqing, Liang, Dan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7705241/
https://www.ncbi.nlm.nih.gov/pubmed/33281814
http://dx.doi.org/10.3389/fimmu.2020.581673
_version_ 1783616919039901696
author Chen, Yuxi
Li, Zhuang
Li, He
Su, Wenru
Xie, Yanyan
Pan, Yuan
Chen, Xiaoqing
Liang, Dan
author_facet Chen, Yuxi
Li, Zhuang
Li, He
Su, Wenru
Xie, Yanyan
Pan, Yuan
Chen, Xiaoqing
Liang, Dan
author_sort Chen, Yuxi
collection PubMed
description Autoimmune uveitis (AU), being one of the sight-threatening ocular inflammatory disorders, has been widely regarded by ophthalmologists and immunologists as a great challenge. Apremilast, a phosphodiesterase-4 inhibitor (PDE4i), which was approved by the U.S. Food and Drug Administration (FDA) for the treatment of active psoriatic arthritis in 2014, has been attracting researchers, who are exploring its efficiency and mechanism on uveitis. In this study, we used an experimental autoimmune uveitis (EAU), a representative model for human AU, to investigate the effect of apremilast on regulating anti-inflammatory mediators. Our study demonstrated that apremilast treatment resulted in a decrease in vascular leakage, macular edema, and inflammatory cell infiltration in the retina, corresponding to decreased clinical and pathological scores. Specifically, apremilast decreased the proportion and population of Th17 cells and increased the proportion and population of T regulatory (Treg) cells. Mechanistically, apremilast may regulate Th17 and Treg cells by inhibiting the phosphorylation of the phosphoinositide 3-kinase (PI3K)/protein kinase B(AKT)/Forkhead box O1 (FoxO1) signaling pathway. These findings suggested that apremilast alleviated EAU by regulating Th17 and Treg through the PI3K/AKT/FoxO1 pathway.
format Online
Article
Text
id pubmed-7705241
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-77052412020-12-03 Apremilast Regulates the Teff/Treg Balance to Ameliorate Uveitis via PI3K/AKT/FoxO1 Signaling Pathway Chen, Yuxi Li, Zhuang Li, He Su, Wenru Xie, Yanyan Pan, Yuan Chen, Xiaoqing Liang, Dan Front Immunol Immunology Autoimmune uveitis (AU), being one of the sight-threatening ocular inflammatory disorders, has been widely regarded by ophthalmologists and immunologists as a great challenge. Apremilast, a phosphodiesterase-4 inhibitor (PDE4i), which was approved by the U.S. Food and Drug Administration (FDA) for the treatment of active psoriatic arthritis in 2014, has been attracting researchers, who are exploring its efficiency and mechanism on uveitis. In this study, we used an experimental autoimmune uveitis (EAU), a representative model for human AU, to investigate the effect of apremilast on regulating anti-inflammatory mediators. Our study demonstrated that apremilast treatment resulted in a decrease in vascular leakage, macular edema, and inflammatory cell infiltration in the retina, corresponding to decreased clinical and pathological scores. Specifically, apremilast decreased the proportion and population of Th17 cells and increased the proportion and population of T regulatory (Treg) cells. Mechanistically, apremilast may regulate Th17 and Treg cells by inhibiting the phosphorylation of the phosphoinositide 3-kinase (PI3K)/protein kinase B(AKT)/Forkhead box O1 (FoxO1) signaling pathway. These findings suggested that apremilast alleviated EAU by regulating Th17 and Treg through the PI3K/AKT/FoxO1 pathway. Frontiers Media S.A. 2020-11-17 /pmc/articles/PMC7705241/ /pubmed/33281814 http://dx.doi.org/10.3389/fimmu.2020.581673 Text en Copyright © 2020 Chen, Li, Li, Su, Xie, Pan, Chen and Liang http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Immunology
Chen, Yuxi
Li, Zhuang
Li, He
Su, Wenru
Xie, Yanyan
Pan, Yuan
Chen, Xiaoqing
Liang, Dan
Apremilast Regulates the Teff/Treg Balance to Ameliorate Uveitis via PI3K/AKT/FoxO1 Signaling Pathway
title Apremilast Regulates the Teff/Treg Balance to Ameliorate Uveitis via PI3K/AKT/FoxO1 Signaling Pathway
title_full Apremilast Regulates the Teff/Treg Balance to Ameliorate Uveitis via PI3K/AKT/FoxO1 Signaling Pathway
title_fullStr Apremilast Regulates the Teff/Treg Balance to Ameliorate Uveitis via PI3K/AKT/FoxO1 Signaling Pathway
title_full_unstemmed Apremilast Regulates the Teff/Treg Balance to Ameliorate Uveitis via PI3K/AKT/FoxO1 Signaling Pathway
title_short Apremilast Regulates the Teff/Treg Balance to Ameliorate Uveitis via PI3K/AKT/FoxO1 Signaling Pathway
title_sort apremilast regulates the teff/treg balance to ameliorate uveitis via pi3k/akt/foxo1 signaling pathway
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7705241/
https://www.ncbi.nlm.nih.gov/pubmed/33281814
http://dx.doi.org/10.3389/fimmu.2020.581673
work_keys_str_mv AT chenyuxi apremilastregulatesthetefftregbalancetoameliorateuveitisviapi3kaktfoxo1signalingpathway
AT lizhuang apremilastregulatesthetefftregbalancetoameliorateuveitisviapi3kaktfoxo1signalingpathway
AT lihe apremilastregulatesthetefftregbalancetoameliorateuveitisviapi3kaktfoxo1signalingpathway
AT suwenru apremilastregulatesthetefftregbalancetoameliorateuveitisviapi3kaktfoxo1signalingpathway
AT xieyanyan apremilastregulatesthetefftregbalancetoameliorateuveitisviapi3kaktfoxo1signalingpathway
AT panyuan apremilastregulatesthetefftregbalancetoameliorateuveitisviapi3kaktfoxo1signalingpathway
AT chenxiaoqing apremilastregulatesthetefftregbalancetoameliorateuveitisviapi3kaktfoxo1signalingpathway
AT liangdan apremilastregulatesthetefftregbalancetoameliorateuveitisviapi3kaktfoxo1signalingpathway