1. Academic Validation
  2. O-GlcNAc transferase couples MRE11 to transcriptionally active chromatin to suppress DNA damage

O-GlcNAc transferase couples MRE11 to transcriptionally active chromatin to suppress DNA damage

  • J Biomed Sci. 2022 Feb 14;29(1):13. doi: 10.1186/s12929-022-00795-1.
Aishwarya Gondane 1 Samuel Girmay 1 Alma Helevä 1 Satu Pallasaho 1 Massimo Loda 2 3 4 Harri M Itkonen 5 6
Affiliations

Affiliations

  • 1 Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
  • 2 Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY, USA.
  • 3 The Broad Institute of Harvard and MIT, Cambridge, MA, USA.
  • 4 The New York Genome Center, New York, NY, USA.
  • 5 Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland. [email protected].
  • 6 Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY, USA. [email protected].
Abstract

Background: Transcription, metabolism and DNA damage response are tightly regulated to preserve the genomic integrity, and O-GlcNAc transferase (OGT) is positioned to connect the three. Prostate Cancer is the most common Cancer in men, and androgen-ablation therapy halts disease progression. However, a significant number of prostate Cancer patients develop resistance against anti-androgens, and this incurable disease is termed castration-resistant prostate Cancer (CRPC). We have shown that combined inhibition of OGT and the transcription elongation kinase CDK9 induce CRPC-selective anti-proliferative effects. Here, we explain the functional basis for these combinatorial effects.

Methods: We used comprehensive mass spectrometry profiling of short-term CDK9 Inhibitor effects on O-GlcNAcylated proteins in an isogenic cell line system that models transition from PC to CRPC. In addition, we used both ChIP-seq and RNA-seq profiling, and pulldown experiments in multiple CRPC models. Finally, we validated our findings in prostate Cancer patient samples.

Results: Inhibition of CDK9 results in an OGT-dependent remodeling of the proteome in prostate Cancer cells. More specifically, the activity of the DNA damage repair protein MRE11 is regulated in response to CDK9 inhibition in an OGT-dependent manner. MRE11 is enriched at the O-GlcNAc-marked loci. CDK9 inhibition does not decrease the expression of mRNAs whose genes are bound by both O-GlcNAc and MRE11. Combined inhibition of CDK9 and OGT or MRE11 further decreases RNA polymerase II activity, induces DNA damage signaling, and blocks the survival of prostate Cancer cells. These effects are seen in CRPC cells but not in normal prostate cells. Mechanistically, OGT activity is required for MRE11 chromatin-loading in cells treated with CDK9 Inhibitor. Finally, we show that MRE11 and O-GlcNAc are enriched at the prostate cancer-specific small nucleotide polymorphic sites, and the loss of MRE11 activity results in a hyper-mutator phenotype in patient tumors.

Conclusions: Both OGT and MRE11 are essential for the repair of CDK9 inhibitor-induced DNA damage. Our study raises the possibility of targeting CDK9 to elicit DNA damage in CRPC setting as an adjuvant to other treatments.

Keywords

Castration-resistant prostate cancer; Cyclin-dependent kinase 9; DNA damage; MRE11; O-GlcNAc transferase.

Figures
Products