Overexpression of HDAC9 promotes oral squamous cell carcinoma growth, regulates cell cycle progression, and inhibits apoptosis

B Rastogi, SK Raut, NK Panda, V Rattan… - Molecular and cellular …, 2016 - Springer
B Rastogi, SK Raut, NK Panda, V Rattan, BD Radotra, M Khullar
Molecular and cellular biochemistry, 2016Springer
Histone deacetylases (HDACs) are a family of deacetylase enzymes that regulate the
acetylation state of histones and a variety of other non-histone proteins including key
oncogenic and tumor suppressor proteins, which modulates chromatin conformation,
leading to regulation of gene expression. HDACs has been grouped into classes I–IV and
histone deacetylase 9 (HDAC9) belongs to class IIa which exhibits tissue-specific
expression. Recent reports have demonstrated both pro-oncogenic and tumor suppressive …
Abstract
Histone deacetylases (HDACs) are a family of deacetylase enzymes that regulate the acetylation state of histones and a variety of other non-histone proteins including key oncogenic and tumor suppressor proteins, which modulates chromatin conformation, leading to regulation of gene expression. HDACs has been grouped into classes I–IV and histone deacetylase 9 (HDAC9) belongs to class IIa which exhibits tissue-specific expression. Recent reports have demonstrated both pro-oncogenic and tumor suppressive role for HDAC9 in different cancers; however, its role in OSCC remains elusive. Here, we investigated the role of HDAC9 in pathogenesis of oral squamous cell carcinoma (OSCC). Our data showed significantly increased mRNA and protein expression of HDAC9 in clinical OSCC samples and UPCI-SCC-116 cells as compared to normal counterpart. Kaplan–Meier analysis showed that the patients with high-level of HDAC9 expression had significantly reduced overall survival than those with low-level of HDAC9 expression (p = 0.034). Knockdown of HDAC9 using siRNA interference suppressed cell proliferation, increased apoptosis, and induced G0/G1 cell cycle arrest in UPCI-SCC-116 cells. Immunofluorescence analysis showed increased nuclear localization of HDAC9 in frozen OSCC sections, and indicative of active HDAC9 that may transcriptionally repress its downstream target genes. Subsequent investigation revealed that overexpression of HDAC9 contributes to OSCC carcinogenesis via targeting a transcription factor, MEF2D, and NR4A1/Nur77, a pro-apoptotic MEF2 target.
Springer