Cargando…

Identification of HDAC9 as a viable therapeutic target for the treatment of gastric cancer

Histone deacetylase inhibitors (HDACis) are a new class of anticancer drugs confirmed to have good therapeutic effects against gastric cancer (GC) in preclinical experiments, but most HDACis are non-selective (pan-HDACis), with highly toxic side effects. Therefore, it is necessary to screen HDAC fam...

Descripción completa

Detalles Bibliográficos
Autores principales: Xiong, Kai, Zhang, Hejun, Du, Yang, Tian, Jie, Ding, Shigang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6802628/
https://www.ncbi.nlm.nih.gov/pubmed/31451695
http://dx.doi.org/10.1038/s12276-019-0301-8
_version_ 1783460824654807040
author Xiong, Kai
Zhang, Hejun
Du, Yang
Tian, Jie
Ding, Shigang
author_facet Xiong, Kai
Zhang, Hejun
Du, Yang
Tian, Jie
Ding, Shigang
author_sort Xiong, Kai
collection PubMed
description Histone deacetylase inhibitors (HDACis) are a new class of anticancer drugs confirmed to have good therapeutic effects against gastric cancer (GC) in preclinical experiments, but most HDACis are non-selective (pan-HDACis), with highly toxic side effects. Therefore, it is necessary to screen HDAC family members that play key roles in GC as therapeutic targets to reduce toxic side effects. In this study, we evaluated the targeting specificity of the HDACi suberoylanilide hydroxamic acid (SAHA) for GC via fluorescence molecular imaging (FMI). In vitro FMI results showed that SAHA had higher binding affinity for GC cells than for normal gastric cells. In vivo FMI of gastric tumor-bearing mice confirmed that SAHA can be enriched in GC tissues. However, there was also a high-concentration distribution in normal organs such as the stomach and lungs, suggesting potential side effects. In addition, we found that among the HDAC family members, HDAC9 was the most significantly upregulated in GC cells, and we verified this upregulation in GC tissues. Further experiments confirmed that knockdown of HDAC9 inhibits cell growth, reduces colony formation, and induces apoptosis and cell cycle arrest. These results suggest that HDAC9 has an oncogenic role in GC. Moreover, HDAC9 siRNA suppressed GC tumor growth and enhanced the antitumor efficacy of cisplatin in GC treatment by inhibiting the proliferation and inducing the apoptosis of GC cells in vitro and in vivo. Our findings suggest that the development of HDAC9-selective HDACis is a potential approach to improve the efficacy of chemotherapy and reduce systemic toxicity.
format Online
Article
Text
id pubmed-6802628
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-68026282019-10-29 Identification of HDAC9 as a viable therapeutic target for the treatment of gastric cancer Xiong, Kai Zhang, Hejun Du, Yang Tian, Jie Ding, Shigang Exp Mol Med Article Histone deacetylase inhibitors (HDACis) are a new class of anticancer drugs confirmed to have good therapeutic effects against gastric cancer (GC) in preclinical experiments, but most HDACis are non-selective (pan-HDACis), with highly toxic side effects. Therefore, it is necessary to screen HDAC family members that play key roles in GC as therapeutic targets to reduce toxic side effects. In this study, we evaluated the targeting specificity of the HDACi suberoylanilide hydroxamic acid (SAHA) for GC via fluorescence molecular imaging (FMI). In vitro FMI results showed that SAHA had higher binding affinity for GC cells than for normal gastric cells. In vivo FMI of gastric tumor-bearing mice confirmed that SAHA can be enriched in GC tissues. However, there was also a high-concentration distribution in normal organs such as the stomach and lungs, suggesting potential side effects. In addition, we found that among the HDAC family members, HDAC9 was the most significantly upregulated in GC cells, and we verified this upregulation in GC tissues. Further experiments confirmed that knockdown of HDAC9 inhibits cell growth, reduces colony formation, and induces apoptosis and cell cycle arrest. These results suggest that HDAC9 has an oncogenic role in GC. Moreover, HDAC9 siRNA suppressed GC tumor growth and enhanced the antitumor efficacy of cisplatin in GC treatment by inhibiting the proliferation and inducing the apoptosis of GC cells in vitro and in vivo. Our findings suggest that the development of HDAC9-selective HDACis is a potential approach to improve the efficacy of chemotherapy and reduce systemic toxicity. Nature Publishing Group UK 2019-08-26 /pmc/articles/PMC6802628/ /pubmed/31451695 http://dx.doi.org/10.1038/s12276-019-0301-8 Text en © The Author(s) 2019 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/.
spellingShingle Article
Xiong, Kai
Zhang, Hejun
Du, Yang
Tian, Jie
Ding, Shigang
Identification of HDAC9 as a viable therapeutic target for the treatment of gastric cancer
title Identification of HDAC9 as a viable therapeutic target for the treatment of gastric cancer
title_full Identification of HDAC9 as a viable therapeutic target for the treatment of gastric cancer
title_fullStr Identification of HDAC9 as a viable therapeutic target for the treatment of gastric cancer
title_full_unstemmed Identification of HDAC9 as a viable therapeutic target for the treatment of gastric cancer
title_short Identification of HDAC9 as a viable therapeutic target for the treatment of gastric cancer
title_sort identification of hdac9 as a viable therapeutic target for the treatment of gastric cancer
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6802628/
https://www.ncbi.nlm.nih.gov/pubmed/31451695
http://dx.doi.org/10.1038/s12276-019-0301-8
work_keys_str_mv AT xiongkai identificationofhdac9asaviabletherapeutictargetforthetreatmentofgastriccancer
AT zhanghejun identificationofhdac9asaviabletherapeutictargetforthetreatmentofgastriccancer
AT duyang identificationofhdac9asaviabletherapeutictargetforthetreatmentofgastriccancer
AT tianjie identificationofhdac9asaviabletherapeutictargetforthetreatmentofgastriccancer
AT dingshigang identificationofhdac9asaviabletherapeutictargetforthetreatmentofgastriccancer