XRCC5 cooperates with p300 to promote cyclooxygenase-2 expression and tumor growth in colon cancers

PLoS One. 2017 Oct 19;12(10):e0186900. doi: 10.1371/journal.pone.0186900. eCollection 2017.

Abstract

Cyclooxygenase (COX) is the rate-limiting enzyme in prostaglandins (PGs) biosynthesis. Previous studies indicate that COX-2, one of the isoforms of COX, is highly expressed in colon cancers and plays a key role in colon cancer carcinogenesis. Thus, searching for novel transcription factors regulating COX-2 expression will facilitate drug development for colon cancer. In this study, we identified XRCC5 as a binding protein of the COX-2 gene promoter in colon cancer cells with streptavidin-agarose pulldown assay and mass spectrometry analysis, and found that XRCC5 promoted colon cancer growth through modulation of COX-2 signaling. Knockdown of XRCC5 by siRNAs inhibited the growth of colon cancer cells in vitro and of tumor xenografts in a mouse model in vivo by suppressing COX-2 promoter activity and COX-2 protein expression. Conversely, overexpression of XRCC5 promoted the growth of colon cancer cells by activating COX-2 promoter and increasing COX-2 protein expression. Moreover, the role of p300 (a transcription co-activator) in acetylating XRCC5 to co-regulate COX-2 expression was also evaluated. Immunofluorescence assay and confocal microscopy showed that XRCC5 and p300 proteins were co-located in the nucleus of colon cancer cells. Co-immunoprecipitation assay also proved the interaction between XRCC5 and p300 in nuclear proteins of colon cancer cells. Cell viability assay indicated that the overexpression of wild-type p300, but not its histone acetyltransferase (HAT) domain deletion mutant, increased XRCC5 acetylation, thereby up-regulated COX-2 expression and promoted the growth of colon cancer cells. In contrast, suppression of p300 by a p300 HAT-specific inhibitor (C646) inhibited colon cancer cell growth by suppressing COX-2 expression. Taken together, our results demonstrated that XRCC5 promoted colon cancer growth by cooperating with p300 to regulate COX-2 expression, and suggested that the XRCC5/p300/COX-2 signaling pathway was a potential target in the treatment of colon cancers.

MeSH terms

  • Animals
  • Cell Line, Tumor
  • Cell Proliferation*
  • Colonic Neoplasms / enzymology
  • Colonic Neoplasms / metabolism*
  • Colonic Neoplasms / pathology
  • Cyclooxygenase 2 / metabolism*
  • Heterografts
  • Humans
  • Ku Autoantigen / metabolism*
  • Mice
  • Protein Binding
  • p300-CBP Transcription Factors / metabolism*

Substances

  • Cyclooxygenase 2
  • p300-CBP Transcription Factors
  • XRCC5 protein, human
  • Ku Autoantigen

Grants and funding

This work was supported by the funds from the National Natural Science Foundation of China (81470337, 81572706, 81472178, 81272195), the Education Department of Liaoning Province, China (‘‘the Program for Pan-Deng Scholars’’), the scientific research project from the Education Department of Liaoning Province, China (L2015142). Guangzhou Double Bioproduct Inc provided some salaries for authors (WH, WD) but did not have any role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript. The specific roles of these authors are articulated in the 'author contributions' section.