Cargando…

Stress-induced nuclear translocation of CDK5 suppresses neuronal death by downregulating ERK activation via VRK3 phosphorylation

Although extracellular signal-related kinase 1/2 (ERK 1/2) activity is generally associated with cell survival, prolonged ERK activation induced by oxidative stress also mediates neuronal cell death. Here we report that oxidative stress-induced cyclin-dependent kinase 5 (CDK5) activation stimulates...

Descripción completa

Detalles Bibliográficos
Autores principales: Song, Haengjin, Kim, Wanil, Choi, Jung-Hyun, Kim, Sung-Hoon, Lee, Dohyun, Park, Choon-Ho, Kim, Sangjune, Kim, Do-Yeon, Kim, Kyong-Tai
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4922050/
https://www.ncbi.nlm.nih.gov/pubmed/27346674
http://dx.doi.org/10.1038/srep28634
_version_ 1782439573768372224
author Song, Haengjin
Kim, Wanil
Choi, Jung-Hyun
Kim, Sung-Hoon
Lee, Dohyun
Park, Choon-Ho
Kim, Sangjune
Kim, Do-Yeon
Kim, Kyong-Tai
author_facet Song, Haengjin
Kim, Wanil
Choi, Jung-Hyun
Kim, Sung-Hoon
Lee, Dohyun
Park, Choon-Ho
Kim, Sangjune
Kim, Do-Yeon
Kim, Kyong-Tai
author_sort Song, Haengjin
collection PubMed
description Although extracellular signal-related kinase 1/2 (ERK 1/2) activity is generally associated with cell survival, prolonged ERK activation induced by oxidative stress also mediates neuronal cell death. Here we report that oxidative stress-induced cyclin-dependent kinase 5 (CDK5) activation stimulates neuroprotective signaling via phosphorylation of vaccinia-related kinase 3 (VRK3) at Ser 108. The binding of vaccinia H1-related (VHR) phosphatase to phosphorylated VRK3 increased its affinity for phospho-ERK and subsequently downregulated ERK activation. Overexpression of VRK3 protected human neuroblastoma SH-SY5Y cells against hydrogen peroxide (H(2)O(2))-induced apoptosis. However the CDK5 was unable to phosphorylate mutant VRK3, and thus the mutant forms of VRK3 could not attenuate apoptotic process. Suppression of CDK5 activity results in increase of ERK activation and elevation of proapoptotic protein Bak expression in mouse cortical neurons. Results from VRK3-deficient neurons were further confirmed the role of VRK3 phosphorylation in H(2)O(2)-evoked ERK regulation. Importantly, we showed an association between phospho-VRK3 levels and the progression of human Alzheimer’s disease (AD) and Parkinson’s disease (PD). Together our work reveals endogenous protective mechanism against oxidative stress-induced neuronal cell death and suggest VRK3 as a potential therapeutic target in neurodegenerative diseases.
format Online
Article
Text
id pubmed-4922050
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Nature Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-49220502016-06-28 Stress-induced nuclear translocation of CDK5 suppresses neuronal death by downregulating ERK activation via VRK3 phosphorylation Song, Haengjin Kim, Wanil Choi, Jung-Hyun Kim, Sung-Hoon Lee, Dohyun Park, Choon-Ho Kim, Sangjune Kim, Do-Yeon Kim, Kyong-Tai Sci Rep Article Although extracellular signal-related kinase 1/2 (ERK 1/2) activity is generally associated with cell survival, prolonged ERK activation induced by oxidative stress also mediates neuronal cell death. Here we report that oxidative stress-induced cyclin-dependent kinase 5 (CDK5) activation stimulates neuroprotective signaling via phosphorylation of vaccinia-related kinase 3 (VRK3) at Ser 108. The binding of vaccinia H1-related (VHR) phosphatase to phosphorylated VRK3 increased its affinity for phospho-ERK and subsequently downregulated ERK activation. Overexpression of VRK3 protected human neuroblastoma SH-SY5Y cells against hydrogen peroxide (H(2)O(2))-induced apoptosis. However the CDK5 was unable to phosphorylate mutant VRK3, and thus the mutant forms of VRK3 could not attenuate apoptotic process. Suppression of CDK5 activity results in increase of ERK activation and elevation of proapoptotic protein Bak expression in mouse cortical neurons. Results from VRK3-deficient neurons were further confirmed the role of VRK3 phosphorylation in H(2)O(2)-evoked ERK regulation. Importantly, we showed an association between phospho-VRK3 levels and the progression of human Alzheimer’s disease (AD) and Parkinson’s disease (PD). Together our work reveals endogenous protective mechanism against oxidative stress-induced neuronal cell death and suggest VRK3 as a potential therapeutic target in neurodegenerative diseases. Nature Publishing Group 2016-06-27 /pmc/articles/PMC4922050/ /pubmed/27346674 http://dx.doi.org/10.1038/srep28634 Text en Copyright © 2016, Macmillan Publishers Limited http://creativecommons.org/licenses/by/4.0/ This work is licensed under a Creative Commons Attribution 4.0 International License. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in the credit line; if the material is not included under the Creative Commons license, users will need to obtain permission from the license holder to reproduce the material. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/
spellingShingle Article
Song, Haengjin
Kim, Wanil
Choi, Jung-Hyun
Kim, Sung-Hoon
Lee, Dohyun
Park, Choon-Ho
Kim, Sangjune
Kim, Do-Yeon
Kim, Kyong-Tai
Stress-induced nuclear translocation of CDK5 suppresses neuronal death by downregulating ERK activation via VRK3 phosphorylation
title Stress-induced nuclear translocation of CDK5 suppresses neuronal death by downregulating ERK activation via VRK3 phosphorylation
title_full Stress-induced nuclear translocation of CDK5 suppresses neuronal death by downregulating ERK activation via VRK3 phosphorylation
title_fullStr Stress-induced nuclear translocation of CDK5 suppresses neuronal death by downregulating ERK activation via VRK3 phosphorylation
title_full_unstemmed Stress-induced nuclear translocation of CDK5 suppresses neuronal death by downregulating ERK activation via VRK3 phosphorylation
title_short Stress-induced nuclear translocation of CDK5 suppresses neuronal death by downregulating ERK activation via VRK3 phosphorylation
title_sort stress-induced nuclear translocation of cdk5 suppresses neuronal death by downregulating erk activation via vrk3 phosphorylation
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4922050/
https://www.ncbi.nlm.nih.gov/pubmed/27346674
http://dx.doi.org/10.1038/srep28634
work_keys_str_mv AT songhaengjin stressinducednucleartranslocationofcdk5suppressesneuronaldeathbydownregulatingerkactivationviavrk3phosphorylation
AT kimwanil stressinducednucleartranslocationofcdk5suppressesneuronaldeathbydownregulatingerkactivationviavrk3phosphorylation
AT choijunghyun stressinducednucleartranslocationofcdk5suppressesneuronaldeathbydownregulatingerkactivationviavrk3phosphorylation
AT kimsunghoon stressinducednucleartranslocationofcdk5suppressesneuronaldeathbydownregulatingerkactivationviavrk3phosphorylation
AT leedohyun stressinducednucleartranslocationofcdk5suppressesneuronaldeathbydownregulatingerkactivationviavrk3phosphorylation
AT parkchoonho stressinducednucleartranslocationofcdk5suppressesneuronaldeathbydownregulatingerkactivationviavrk3phosphorylation
AT kimsangjune stressinducednucleartranslocationofcdk5suppressesneuronaldeathbydownregulatingerkactivationviavrk3phosphorylation
AT kimdoyeon stressinducednucleartranslocationofcdk5suppressesneuronaldeathbydownregulatingerkactivationviavrk3phosphorylation
AT kimkyongtai stressinducednucleartranslocationofcdk5suppressesneuronaldeathbydownregulatingerkactivationviavrk3phosphorylation