Cargando…

Sulforaphane metabolites reduce resistance to paclitaxel via microtubule disruption

Long treatment with paclitaxel (PTX) might increase resistance and side-effects causing a failure in cancer chemotherapy. Here we uncovered that either sulforaphane-cysteine (SFN-Cys) or sulforaphane-N-acetyl-cysteine (SFN-NAC) induced apoptosis via phosphorylated ERK1/2-mediated upregulation of 26 ...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Yalin, Zhou, Yan, Zheng, Zhongnan, Li, Juntao, Yan, Yuting, Wu, Wei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6235886/
https://www.ncbi.nlm.nih.gov/pubmed/30429459
http://dx.doi.org/10.1038/s41419-018-1174-9
_version_ 1783370929783439360
author Wang, Yalin
Zhou, Yan
Zheng, Zhongnan
Li, Juntao
Yan, Yuting
Wu, Wei
author_facet Wang, Yalin
Zhou, Yan
Zheng, Zhongnan
Li, Juntao
Yan, Yuting
Wu, Wei
author_sort Wang, Yalin
collection PubMed
description Long treatment with paclitaxel (PTX) might increase resistance and side-effects causing a failure in cancer chemotherapy. Here we uncovered that either sulforaphane-cysteine (SFN-Cys) or sulforaphane-N-acetyl-cysteine (SFN-NAC) induced apoptosis via phosphorylated ERK1/2-mediated upregulation of 26 S proteasome and Hsp70, and downregulation of βIII-tubulin, XIAP, Tau, Stathmin1 and α-tubulin causing microtubule disruption in human PTX-resistant non-small cell lung cancer (NSCLC) cells. Knockdown of either βIII-tubulin or α-tubulin via siRNA increased cell sensitivity to PTX, indicating that these two proteins help cells increase the resistance. Tissue microarray analysis showed that overexpression of βIII-tubulin correlated to NSCLC malignant grading. Immunofluorescence staining also showed that SFN metabolites induced a nest-like microtubule protein distribution with aggregation and disruption. Co-immunoprecipitation showed that SFN metabolites reduced the interaction between βIII-tubulin and Tau, and that between α-tubulin and XIAP. The combination of PTX with SFN metabolites decreased the resistance to PTX, and doses of both PTX and SFN metabolites, and enhanced apoptosis resulting from activated Caspase-3-caused microtubule degradation. Importantly, the effective dose of SFN metabolites combined with 20 nM PTX will be low to 4 μM. Thus, we might combine SFN metabolites with PTX for preclinical trial. Normally, more than 20 μM SFN metabolites only leading to apoptosis for SFN metabolites hindered their applications. These findings will help us develop a low-resistance and high-efficiency chemotherapy via PTX/SFN metabolites combination.
format Online
Article
Text
id pubmed-6235886
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-62358862018-11-15 Sulforaphane metabolites reduce resistance to paclitaxel via microtubule disruption Wang, Yalin Zhou, Yan Zheng, Zhongnan Li, Juntao Yan, Yuting Wu, Wei Cell Death Dis Article Long treatment with paclitaxel (PTX) might increase resistance and side-effects causing a failure in cancer chemotherapy. Here we uncovered that either sulforaphane-cysteine (SFN-Cys) or sulforaphane-N-acetyl-cysteine (SFN-NAC) induced apoptosis via phosphorylated ERK1/2-mediated upregulation of 26 S proteasome and Hsp70, and downregulation of βIII-tubulin, XIAP, Tau, Stathmin1 and α-tubulin causing microtubule disruption in human PTX-resistant non-small cell lung cancer (NSCLC) cells. Knockdown of either βIII-tubulin or α-tubulin via siRNA increased cell sensitivity to PTX, indicating that these two proteins help cells increase the resistance. Tissue microarray analysis showed that overexpression of βIII-tubulin correlated to NSCLC malignant grading. Immunofluorescence staining also showed that SFN metabolites induced a nest-like microtubule protein distribution with aggregation and disruption. Co-immunoprecipitation showed that SFN metabolites reduced the interaction between βIII-tubulin and Tau, and that between α-tubulin and XIAP. The combination of PTX with SFN metabolites decreased the resistance to PTX, and doses of both PTX and SFN metabolites, and enhanced apoptosis resulting from activated Caspase-3-caused microtubule degradation. Importantly, the effective dose of SFN metabolites combined with 20 nM PTX will be low to 4 μM. Thus, we might combine SFN metabolites with PTX for preclinical trial. Normally, more than 20 μM SFN metabolites only leading to apoptosis for SFN metabolites hindered their applications. These findings will help us develop a low-resistance and high-efficiency chemotherapy via PTX/SFN metabolites combination. Nature Publishing Group UK 2018-11-14 /pmc/articles/PMC6235886/ /pubmed/30429459 http://dx.doi.org/10.1038/s41419-018-1174-9 Text en © The Author(s) 2018 https://creativecommons.org/licenses/by/4.0/Open Access This 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 license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license 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 license, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Article
Wang, Yalin
Zhou, Yan
Zheng, Zhongnan
Li, Juntao
Yan, Yuting
Wu, Wei
Sulforaphane metabolites reduce resistance to paclitaxel via microtubule disruption
title Sulforaphane metabolites reduce resistance to paclitaxel via microtubule disruption
title_full Sulforaphane metabolites reduce resistance to paclitaxel via microtubule disruption
title_fullStr Sulforaphane metabolites reduce resistance to paclitaxel via microtubule disruption
title_full_unstemmed Sulforaphane metabolites reduce resistance to paclitaxel via microtubule disruption
title_short Sulforaphane metabolites reduce resistance to paclitaxel via microtubule disruption
title_sort sulforaphane metabolites reduce resistance to paclitaxel via microtubule disruption
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6235886/
https://www.ncbi.nlm.nih.gov/pubmed/30429459
http://dx.doi.org/10.1038/s41419-018-1174-9
work_keys_str_mv AT wangyalin sulforaphanemetabolitesreduceresistancetopaclitaxelviamicrotubuledisruption
AT zhouyan sulforaphanemetabolitesreduceresistancetopaclitaxelviamicrotubuledisruption
AT zhengzhongnan sulforaphanemetabolitesreduceresistancetopaclitaxelviamicrotubuledisruption
AT lijuntao sulforaphanemetabolitesreduceresistancetopaclitaxelviamicrotubuledisruption
AT yanyuting sulforaphanemetabolitesreduceresistancetopaclitaxelviamicrotubuledisruption
AT wuwei sulforaphanemetabolitesreduceresistancetopaclitaxelviamicrotubuledisruption