Cargando…

Ral A, via activating the mitotic checkpoint, sensitizes cells lacking a functional Nf1 to apoptosis in the absence of protein kinase C

Nf1 mutations or deletions are suggested to underlie the tumor predisposition of NF1 (neurofibromatosis type 1) and few treatments are available for treating NF1 patients with advanced malignant tumors. Aberrant activation of Ras in Nf1-deficient conditions is responsible for the promotion of tumori...

Descripción completa

Detalles Bibliográficos
Autores principales: Ganapathy, Suthakar, Fagman, Johan B, Shen, Ling, Yu, Tianqi, Zhou, Xiaodong, Dai, Wei, Makriyannis, Alexandros, Chen, Changyan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals LLC 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5356664/
https://www.ncbi.nlm.nih.gov/pubmed/27741517
http://dx.doi.org/10.18632/oncotarget.12607
_version_ 1782515886284865536
author Ganapathy, Suthakar
Fagman, Johan B
Shen, Ling
Yu, Tianqi
Zhou, Xiaodong
Dai, Wei
Makriyannis, Alexandros
Chen, Changyan
author_facet Ganapathy, Suthakar
Fagman, Johan B
Shen, Ling
Yu, Tianqi
Zhou, Xiaodong
Dai, Wei
Makriyannis, Alexandros
Chen, Changyan
author_sort Ganapathy, Suthakar
collection PubMed
description Nf1 mutations or deletions are suggested to underlie the tumor predisposition of NF1 (neurofibromatosis type 1) and few treatments are available for treating NF1 patients with advanced malignant tumors. Aberrant activation of Ras in Nf1-deficient conditions is responsible for the promotion of tumorigenesis in NF1. PKC is proven to be an important factor in supporting the viability of Nf1-defected cells, but the molecular mechanisms are not fully understood. In this study, we demonstrate that the inhibition of protein kinase C (PKC) by 1-O-Hexadecyl-2-O-methyl-rac-glycerol (HMG, a PKC inhibitor) preferentially sensitizes Nf1-defected cells to apoptosis, via triggering a persistent mitotic arrest. In this process, Ral A is activated. Subsequently, Chk1 is phosphorylated and translocated to the nucleus. Silencing Ral A significantly blocks Chk1 nuclear translocation and releases HMG-treated Nf1-deficient cells from mitotic arrest, resulting in the reduction of the magnitude of apoptosis. Thus, our study reveals that PKC is able to maintain the homeostasis or viability of Nf1-defected cells and may serve as a potential target for developing new therapeutic strategies.
format Online
Article
Text
id pubmed-5356664
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Impact Journals LLC
record_format MEDLINE/PubMed
spelling pubmed-53566642017-04-26 Ral A, via activating the mitotic checkpoint, sensitizes cells lacking a functional Nf1 to apoptosis in the absence of protein kinase C Ganapathy, Suthakar Fagman, Johan B Shen, Ling Yu, Tianqi Zhou, Xiaodong Dai, Wei Makriyannis, Alexandros Chen, Changyan Oncotarget Research Paper Nf1 mutations or deletions are suggested to underlie the tumor predisposition of NF1 (neurofibromatosis type 1) and few treatments are available for treating NF1 patients with advanced malignant tumors. Aberrant activation of Ras in Nf1-deficient conditions is responsible for the promotion of tumorigenesis in NF1. PKC is proven to be an important factor in supporting the viability of Nf1-defected cells, but the molecular mechanisms are not fully understood. In this study, we demonstrate that the inhibition of protein kinase C (PKC) by 1-O-Hexadecyl-2-O-methyl-rac-glycerol (HMG, a PKC inhibitor) preferentially sensitizes Nf1-defected cells to apoptosis, via triggering a persistent mitotic arrest. In this process, Ral A is activated. Subsequently, Chk1 is phosphorylated and translocated to the nucleus. Silencing Ral A significantly blocks Chk1 nuclear translocation and releases HMG-treated Nf1-deficient cells from mitotic arrest, resulting in the reduction of the magnitude of apoptosis. Thus, our study reveals that PKC is able to maintain the homeostasis or viability of Nf1-defected cells and may serve as a potential target for developing new therapeutic strategies. Impact Journals LLC 2016-10-12 /pmc/articles/PMC5356664/ /pubmed/27741517 http://dx.doi.org/10.18632/oncotarget.12607 Text en Copyright: © 2016 Ganapathy et al. http://creativecommons.org/licenses/by/3.0/ 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 credited.
spellingShingle Research Paper
Ganapathy, Suthakar
Fagman, Johan B
Shen, Ling
Yu, Tianqi
Zhou, Xiaodong
Dai, Wei
Makriyannis, Alexandros
Chen, Changyan
Ral A, via activating the mitotic checkpoint, sensitizes cells lacking a functional Nf1 to apoptosis in the absence of protein kinase C
title Ral A, via activating the mitotic checkpoint, sensitizes cells lacking a functional Nf1 to apoptosis in the absence of protein kinase C
title_full Ral A, via activating the mitotic checkpoint, sensitizes cells lacking a functional Nf1 to apoptosis in the absence of protein kinase C
title_fullStr Ral A, via activating the mitotic checkpoint, sensitizes cells lacking a functional Nf1 to apoptosis in the absence of protein kinase C
title_full_unstemmed Ral A, via activating the mitotic checkpoint, sensitizes cells lacking a functional Nf1 to apoptosis in the absence of protein kinase C
title_short Ral A, via activating the mitotic checkpoint, sensitizes cells lacking a functional Nf1 to apoptosis in the absence of protein kinase C
title_sort ral a, via activating the mitotic checkpoint, sensitizes cells lacking a functional nf1 to apoptosis in the absence of protein kinase c
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5356664/
https://www.ncbi.nlm.nih.gov/pubmed/27741517
http://dx.doi.org/10.18632/oncotarget.12607
work_keys_str_mv AT ganapathysuthakar ralaviaactivatingthemitoticcheckpointsensitizescellslackingafunctionalnf1toapoptosisintheabsenceofproteinkinasec
AT fagmanjohanb ralaviaactivatingthemitoticcheckpointsensitizescellslackingafunctionalnf1toapoptosisintheabsenceofproteinkinasec
AT shenling ralaviaactivatingthemitoticcheckpointsensitizescellslackingafunctionalnf1toapoptosisintheabsenceofproteinkinasec
AT yutianqi ralaviaactivatingthemitoticcheckpointsensitizescellslackingafunctionalnf1toapoptosisintheabsenceofproteinkinasec
AT zhouxiaodong ralaviaactivatingthemitoticcheckpointsensitizescellslackingafunctionalnf1toapoptosisintheabsenceofproteinkinasec
AT daiwei ralaviaactivatingthemitoticcheckpointsensitizescellslackingafunctionalnf1toapoptosisintheabsenceofproteinkinasec
AT makriyannisalexandros ralaviaactivatingthemitoticcheckpointsensitizescellslackingafunctionalnf1toapoptosisintheabsenceofproteinkinasec
AT chenchangyan ralaviaactivatingthemitoticcheckpointsensitizescellslackingafunctionalnf1toapoptosisintheabsenceofproteinkinasec