Cargando…

Upregulation of heat shock protein 70 and the differential protein expression induced by tumor necrosis factor-alpha enhances migration and inhibits apoptosis of hepatocellular carcinoma cell HepG2

Tumor necrosis factor alpha (TNFα) plays diverse roles in liver damage and hepatocarcinogenesis with its multipotent bioactivity. However, the influence of TNFα on protein expression of hepatocellular carcinoma (HCC) is incompletely understood. Therefore, we aimed to investigate the differential pro...

Descripción completa

Detalles Bibliográficos
Autores principales: Huang, Bee-Piao, Lin, Chun-Shiang, Wang, Chau-Jong, Kao, Shao-Hsuan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5370291/
https://www.ncbi.nlm.nih.gov/pubmed/28367089
http://dx.doi.org/10.7150/ijms.17861
_version_ 1782518210055110656
author Huang, Bee-Piao
Lin, Chun-Shiang
Wang, Chau-Jong
Kao, Shao-Hsuan
author_facet Huang, Bee-Piao
Lin, Chun-Shiang
Wang, Chau-Jong
Kao, Shao-Hsuan
author_sort Huang, Bee-Piao
collection PubMed
description Tumor necrosis factor alpha (TNFα) plays diverse roles in liver damage and hepatocarcinogenesis with its multipotent bioactivity. However, the influence of TNFα on protein expression of hepatocellular carcinoma (HCC) is incompletely understood. Therefore, we aimed to investigate the differential protein expression of HCC in response to TNFα stimulus. We observed that HepG2 cell revealed a higher resistance to TNFα-induced apoptosis as compared to the non-tumorigenic hepatocyte THLE-2. By using a label-free quantitative proteomic analysis, we found that 520 proteins were differentially expressed in the HepG2 cells exposed to TNFα, including 211 up-regulated and 309 down-regulated proteins. We further confirmed several proteins with significant expression change (TNFα/control ratio>2.0 or <0.5) by immunoblotting using specific antibodies. We also analyzed the differential expressed proteins using Gene ontology and KEGG annotations, and the results implicated that TNFα might regulate ribosome, spliceosome, antigen processing and presentation, and energy metabolism in HepG2 cells. Moreover, we demonstrated that upregulation of heat shock protein 70 (HSP70) was involved in both the promoted migration and the inhibited apoptosis of HepG2 cells in response to TNFα. Collectively, these findings indicate that TNFα alters protein expression such as HSP70, which triggering specific molecular processes and signaling cascades that promote migration and inhibit apoptosis of HepG2 cells.
format Online
Article
Text
id pubmed-5370291
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-53702912017-03-31 Upregulation of heat shock protein 70 and the differential protein expression induced by tumor necrosis factor-alpha enhances migration and inhibits apoptosis of hepatocellular carcinoma cell HepG2 Huang, Bee-Piao Lin, Chun-Shiang Wang, Chau-Jong Kao, Shao-Hsuan Int J Med Sci Research Paper Tumor necrosis factor alpha (TNFα) plays diverse roles in liver damage and hepatocarcinogenesis with its multipotent bioactivity. However, the influence of TNFα on protein expression of hepatocellular carcinoma (HCC) is incompletely understood. Therefore, we aimed to investigate the differential protein expression of HCC in response to TNFα stimulus. We observed that HepG2 cell revealed a higher resistance to TNFα-induced apoptosis as compared to the non-tumorigenic hepatocyte THLE-2. By using a label-free quantitative proteomic analysis, we found that 520 proteins were differentially expressed in the HepG2 cells exposed to TNFα, including 211 up-regulated and 309 down-regulated proteins. We further confirmed several proteins with significant expression change (TNFα/control ratio>2.0 or <0.5) by immunoblotting using specific antibodies. We also analyzed the differential expressed proteins using Gene ontology and KEGG annotations, and the results implicated that TNFα might regulate ribosome, spliceosome, antigen processing and presentation, and energy metabolism in HepG2 cells. Moreover, we demonstrated that upregulation of heat shock protein 70 (HSP70) was involved in both the promoted migration and the inhibited apoptosis of HepG2 cells in response to TNFα. Collectively, these findings indicate that TNFα alters protein expression such as HSP70, which triggering specific molecular processes and signaling cascades that promote migration and inhibit apoptosis of HepG2 cells. Ivyspring International Publisher 2017-02-25 /pmc/articles/PMC5370291/ /pubmed/28367089 http://dx.doi.org/10.7150/ijms.17861 Text en © Ivyspring International Publisher This is an open access article distributed under the terms of the Creative Commons Attribution (CC BY-NC) license (https://creativecommons.org/licenses/by-nc/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Huang, Bee-Piao
Lin, Chun-Shiang
Wang, Chau-Jong
Kao, Shao-Hsuan
Upregulation of heat shock protein 70 and the differential protein expression induced by tumor necrosis factor-alpha enhances migration and inhibits apoptosis of hepatocellular carcinoma cell HepG2
title Upregulation of heat shock protein 70 and the differential protein expression induced by tumor necrosis factor-alpha enhances migration and inhibits apoptosis of hepatocellular carcinoma cell HepG2
title_full Upregulation of heat shock protein 70 and the differential protein expression induced by tumor necrosis factor-alpha enhances migration and inhibits apoptosis of hepatocellular carcinoma cell HepG2
title_fullStr Upregulation of heat shock protein 70 and the differential protein expression induced by tumor necrosis factor-alpha enhances migration and inhibits apoptosis of hepatocellular carcinoma cell HepG2
title_full_unstemmed Upregulation of heat shock protein 70 and the differential protein expression induced by tumor necrosis factor-alpha enhances migration and inhibits apoptosis of hepatocellular carcinoma cell HepG2
title_short Upregulation of heat shock protein 70 and the differential protein expression induced by tumor necrosis factor-alpha enhances migration and inhibits apoptosis of hepatocellular carcinoma cell HepG2
title_sort upregulation of heat shock protein 70 and the differential protein expression induced by tumor necrosis factor-alpha enhances migration and inhibits apoptosis of hepatocellular carcinoma cell hepg2
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5370291/
https://www.ncbi.nlm.nih.gov/pubmed/28367089
http://dx.doi.org/10.7150/ijms.17861
work_keys_str_mv AT huangbeepiao upregulationofheatshockprotein70andthedifferentialproteinexpressioninducedbytumornecrosisfactoralphaenhancesmigrationandinhibitsapoptosisofhepatocellularcarcinomacellhepg2
AT linchunshiang upregulationofheatshockprotein70andthedifferentialproteinexpressioninducedbytumornecrosisfactoralphaenhancesmigrationandinhibitsapoptosisofhepatocellularcarcinomacellhepg2
AT wangchaujong upregulationofheatshockprotein70andthedifferentialproteinexpressioninducedbytumornecrosisfactoralphaenhancesmigrationandinhibitsapoptosisofhepatocellularcarcinomacellhepg2
AT kaoshaohsuan upregulationofheatshockprotein70andthedifferentialproteinexpressioninducedbytumornecrosisfactoralphaenhancesmigrationandinhibitsapoptosisofhepatocellularcarcinomacellhepg2