Cargando…

Regulation of Autophagy Via PERK-eIF2α Effectively Relieve the Radiation Myelitis Induced by Iodine-125

Radiation myelitis is the most serious complication in clinical radiotherapy for spinal metastases. We previously showed that (125)I brachytherapy induced apoptosis of spinal cord neurons accompanied by autophagy. In this study, we further investigated the mechanism by which (125)I radiation trigger...

Descripción completa

Detalles Bibliográficos
Autores principales: Yang, Zuozhang, Xu, Yongqing, Xu, Lei, Maccauro, Giulio, Rossi, Barbara, Chen, Yanjin, Li, Hongjun, Zhang, Jing, Sun, Hongpu, Yang, Yihao, Xu, Da, Liu, Xuefeng
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2013
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3818370/
https://www.ncbi.nlm.nih.gov/pubmed/24223705
http://dx.doi.org/10.1371/journal.pone.0076819
_version_ 1782478174903336960
author Yang, Zuozhang
Xu, Yongqing
Xu, Lei
Maccauro, Giulio
Rossi, Barbara
Chen, Yanjin
Li, Hongjun
Zhang, Jing
Sun, Hongpu
Yang, Yihao
Xu, Da
Liu, Xuefeng
author_facet Yang, Zuozhang
Xu, Yongqing
Xu, Lei
Maccauro, Giulio
Rossi, Barbara
Chen, Yanjin
Li, Hongjun
Zhang, Jing
Sun, Hongpu
Yang, Yihao
Xu, Da
Liu, Xuefeng
author_sort Yang, Zuozhang
collection PubMed
description Radiation myelitis is the most serious complication in clinical radiotherapy for spinal metastases. We previously showed that (125)I brachytherapy induced apoptosis of spinal cord neurons accompanied by autophagy. In this study, we further investigated the mechanism by which (125)I radiation triggered autophagy in neural cells. We found that autophagy induced by (125)I radiation was involved in endoplasmic reticulum (ER) stress and mainly dependent on PERK-eIF2α pathway. The expressions of LC3II, ATG12 and PI3K were significantly suppressed in PERK knockout neural cells. Meanwhile, the expressions of phosphorylated-Akt s473 and caspase3/8 all significantly increased in neural cells transfected with a PERK siRNA and which enhanced apoptosis of neurons after (125)I radiation. The results were consistent with that by MTT and Annexin-FITC/PT staining. In annimal model of banna pigs with radiation myelitis caused by (125)I brachytherapy, we have successfully decreased PERK expression by intrathecal administration of the lentivirus vector. The apoptosis rate was significantly higher than that in control group and which deteriorated radiation myelitis of banna pigs. Thus, autophagy caused by (125)I radiation was mainly as an attempt of cell survival at an early stage, but it would be a self-destructive process and promoted the process of apoptosis and necrosis radiated by (125)I for more than 72 hours. The study would be useful and helpful to maximize efficiency of radiation therapy in clinical therapy.
format Online
Article
Text
id pubmed-3818370
institution National Center for Biotechnology Information
language English
publishDate 2013
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-38183702013-11-09 Regulation of Autophagy Via PERK-eIF2α Effectively Relieve the Radiation Myelitis Induced by Iodine-125 Yang, Zuozhang Xu, Yongqing Xu, Lei Maccauro, Giulio Rossi, Barbara Chen, Yanjin Li, Hongjun Zhang, Jing Sun, Hongpu Yang, Yihao Xu, Da Liu, Xuefeng PLoS One Research Article Radiation myelitis is the most serious complication in clinical radiotherapy for spinal metastases. We previously showed that (125)I brachytherapy induced apoptosis of spinal cord neurons accompanied by autophagy. In this study, we further investigated the mechanism by which (125)I radiation triggered autophagy in neural cells. We found that autophagy induced by (125)I radiation was involved in endoplasmic reticulum (ER) stress and mainly dependent on PERK-eIF2α pathway. The expressions of LC3II, ATG12 and PI3K were significantly suppressed in PERK knockout neural cells. Meanwhile, the expressions of phosphorylated-Akt s473 and caspase3/8 all significantly increased in neural cells transfected with a PERK siRNA and which enhanced apoptosis of neurons after (125)I radiation. The results were consistent with that by MTT and Annexin-FITC/PT staining. In annimal model of banna pigs with radiation myelitis caused by (125)I brachytherapy, we have successfully decreased PERK expression by intrathecal administration of the lentivirus vector. The apoptosis rate was significantly higher than that in control group and which deteriorated radiation myelitis of banna pigs. Thus, autophagy caused by (125)I radiation was mainly as an attempt of cell survival at an early stage, but it would be a self-destructive process and promoted the process of apoptosis and necrosis radiated by (125)I for more than 72 hours. The study would be useful and helpful to maximize efficiency of radiation therapy in clinical therapy. Public Library of Science 2013-11-05 /pmc/articles/PMC3818370/ /pubmed/24223705 http://dx.doi.org/10.1371/journal.pone.0076819 Text en © 2013 Yang et al http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Yang, Zuozhang
Xu, Yongqing
Xu, Lei
Maccauro, Giulio
Rossi, Barbara
Chen, Yanjin
Li, Hongjun
Zhang, Jing
Sun, Hongpu
Yang, Yihao
Xu, Da
Liu, Xuefeng
Regulation of Autophagy Via PERK-eIF2α Effectively Relieve the Radiation Myelitis Induced by Iodine-125
title Regulation of Autophagy Via PERK-eIF2α Effectively Relieve the Radiation Myelitis Induced by Iodine-125
title_full Regulation of Autophagy Via PERK-eIF2α Effectively Relieve the Radiation Myelitis Induced by Iodine-125
title_fullStr Regulation of Autophagy Via PERK-eIF2α Effectively Relieve the Radiation Myelitis Induced by Iodine-125
title_full_unstemmed Regulation of Autophagy Via PERK-eIF2α Effectively Relieve the Radiation Myelitis Induced by Iodine-125
title_short Regulation of Autophagy Via PERK-eIF2α Effectively Relieve the Radiation Myelitis Induced by Iodine-125
title_sort regulation of autophagy via perk-eif2α effectively relieve the radiation myelitis induced by iodine-125
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3818370/
https://www.ncbi.nlm.nih.gov/pubmed/24223705
http://dx.doi.org/10.1371/journal.pone.0076819
work_keys_str_mv AT yangzuozhang regulationofautophagyviaperkeif2aeffectivelyrelievetheradiationmyelitisinducedbyiodine125
AT xuyongqing regulationofautophagyviaperkeif2aeffectivelyrelievetheradiationmyelitisinducedbyiodine125
AT xulei regulationofautophagyviaperkeif2aeffectivelyrelievetheradiationmyelitisinducedbyiodine125
AT maccaurogiulio regulationofautophagyviaperkeif2aeffectivelyrelievetheradiationmyelitisinducedbyiodine125
AT rossibarbara regulationofautophagyviaperkeif2aeffectivelyrelievetheradiationmyelitisinducedbyiodine125
AT chenyanjin regulationofautophagyviaperkeif2aeffectivelyrelievetheradiationmyelitisinducedbyiodine125
AT lihongjun regulationofautophagyviaperkeif2aeffectivelyrelievetheradiationmyelitisinducedbyiodine125
AT zhangjing regulationofautophagyviaperkeif2aeffectivelyrelievetheradiationmyelitisinducedbyiodine125
AT sunhongpu regulationofautophagyviaperkeif2aeffectivelyrelievetheradiationmyelitisinducedbyiodine125
AT yangyihao regulationofautophagyviaperkeif2aeffectivelyrelievetheradiationmyelitisinducedbyiodine125
AT xuda regulationofautophagyviaperkeif2aeffectivelyrelievetheradiationmyelitisinducedbyiodine125
AT liuxuefeng regulationofautophagyviaperkeif2aeffectivelyrelievetheradiationmyelitisinducedbyiodine125