Elsevier

Leukemia Research

Volume 45, June 2016, Pages 14-23
Leukemia Research

Histone deacetylase inhibitors decrease NHEJ both by acetylation of repair factors and trapping of PARP1 at DNA double-strand breaks in chromatin

https://doi.org/10.1016/j.leukres.2016.03.007Get rights and content

Highlights

Abstract

Histone deacetylase inhibitors (HDACi) induce acetylation of histone and non-histone proteins, and modulate the acetylation of proteins involved in DNA double-strand break (DSB) repair. Non-homologous end-joining (NHEJ) is one of the main pathways for repairing DSBs. Decreased NHEJ activity has been reported with HDACi treatment. However, mechanisms through which these effects are regulated in the context of chromatin are unclear. We show that pan-HDACi, trichostatin A (TSA), causes differential acetylation of DNA repair factors Ku70/Ku80 and poly ADP-ribose polymerase-1 (PARP1), and impairs NHEJ. Repair effects are reversed by treatments with p300/CBP inhibitor C646, with significantly decreased acetylation of PARP1. In keeping with these findings, TSA treatment significantly increases PARP1 binding to DSBs in chromatin. Notably, AML patients treated with HDACi entinostat (MS275) in vivo also show increased formation of poly ADP-ribose (PAR) that co-localizes with DSBs. Further, we demonstrate that PARP1 bound to chromatin increases with duration of TSA exposure, resembling PARP “trapping”. Knockdown of PARP1 inhibits trapping and mitigates HDACi effects on NHEJ. Finally, combination of HDACi with potent PARP inhibitor talazoparib (BMN673) shows a dose-dependent increase in PARP “trapping”, which correlates with increased apoptosis. These results provide a mechanism through which HDACi inhibits deacetylation and increases binding of PARP1 to DSBs, leading to decreased NHEJ and cytotoxicity of leukemia cells.

Introduction

Histone deacetylases (HDACs) catalyze the removal of acetyl groups from the amino-terminal lysine residues of histone proteins, while histone acetyltransferases (HATs) promote their addition, leading to local remodeling of chromatin, a critical step for the access of regulatory proteins to DNA [1]. In addition to their activity on histone proteins, HDACs and HATs also target non-histone proteins. Transcription factors, transcription regulators and DNA repair proteins have been shown to be directly or indirectly regulated by HDACs and HAT enzymes [2]. In leukemias, specific oncogenic fusion proteins recruit HDACs to the promoters of their target genes. These events appear to contribute to leukemogenesis [3], suggesting that these diseases are particularly good candidates for treatment with HDAC inhibitors (HDACi) [4]. Indeed, clinical trials of HDACi in hematological malignancies, such as lymphoma [5] and myelodysplastic syndromes [6] are yielding promising results. Recently, U.S. Food and Drug Administration (FDA) approved HDACi panobinostat for treatment of multiple myeloma [7]. Also, Vorinostat and Romidepsin received FDA approval for treatment of cutaneous T-cell lymphoma [8], [9], [10].

Although HDACi treatment results in apoptosis of cancer cells in vivo, the mechanism underlying this effect is still unclear. It has been reported that HDACi induce apoptosis by upregulation of both death receptors and their ligands in leukemia cells [11], [12], and by activation of the mitochondrial/intrinsic apoptotic pathway through selective activation of Bid and induction of reactive oxygen species (ROS) [13]. In addition, we and others have shown that HDACi cause DNA damage that is significantly increased in solid tumors [14] and leukemias [15] compared with normal cells. Increases in γH2AX and ATM phosphorylation, that are early indicators of DNA damage [16], [17], occur rapidly following HDACi administration [15]. Furthermore, treatment with HDACi such as trichostatin A (TSA) leads to a persistence of DNA double strand breaks (DSBs) [14], [15], and sensitize solid tumor and leukemia cells to ionizing radiation [14], [18], [19], [20].

There are two major mechanisms for the repair of DSBs [21], homologous recombination (HR) and the classical DNA-dependent protein kinase (DNA-PKcs)-dependent non-homologous end-joining (c-NHEJ or NHEJ). Recent studies have also identified an alternative and highly error-prone repair pathway, Alt-NHEJ [22], [23], [24] that appears upregulated in cancer and leukemia cells [25], and is implicated as a mechanism for genomic instability [22], [23], [24]. Poly ADP-ribose polymerase-1 (PARP1), that participates in several DNA repair pathways, including single strand break (SSB) repair, is also a key player for initiation of Alt-NHEJ [26].

Several studies have investigated mechanisms by which HDACi modulate DSB repair [27], [28], [29], [30]. Mouse embryonic fibroblasts mutated for NHEJ proteins Ku80 or DNA Ligase IV (LigIV), but not for the HR protein BRCA2, are hypersensitive to TSA [31]. In human prostate cancer cells, HDACi cause hyperacetylation of Ku proteins and decrease the binding of these proteins to DNA [32]. Vorinostat, which is also a pan-HDACi like TSA, downregulates DNA-PKcs in melanoma cells [14]. We previously observed increased co-localization of DNA-PKcs and γH2AX in HDACi-treated cells indicating that NHEJ components localize in vicinity of DSBs after drug treatment [15]. However, acetylation of Ku70 by HDACi decreases overall NHEJ activity [27], [28], [29]. Thus, it is unclear whether repair factors assembled around DSBs in chromatin following HDAC inhibition are able to access DSBs and perform repair. In this study, we investigated whether HDACi impact recruitment of DSB repair proteins to chromatin, particularly those involved in NHEJ.

We show that pan-HDACi TSA treatment of acute leukemia cells results in differential acetylation of Ku70, Ku80, and PARP1, leading to decreased NHEJ activity. We also show that HAT p300/CBP inhibition using the pharmacological inhibitor C646 reverses these effects, specifically affecting PARP1 acetylation. Moreover, PARP1 is significantly enriched in chromatin following HDACi, compared with Ku proteins, in a mechanism similar to PARP1 trapping observed with PARP inhibitors (PARPis). Notably, PARPis in combination with HDACi lead to further increases in PARP trapping that correlates with increased apoptosis. This suggests that HDACi result in both a physical and functional alteration of PARP1 binding at DSBs, potentially preventing access of NHEJ factors, and therefore resulting in decreased NHEJ repair.

Section snippets

Cell culture, patients and treatment

K562 and HL60 cell lines from American Type Culture Collection (ATCC, Manassas, VA) were grown in RPMI 1640 medium with 25 mM HEPES (Cellgro, Mediatech Inc., Manassas, VA) supplemented with 2 mM glutamine (Cellgro), 1% penicillin/streptomycin (Gibco, Invitrogen, Carlsbad, CA) and 10% fetal bovine serum (FBS, ATCC) at 37 °C and 5% CO2. A derivative of K562 that stably expresses DRneo construct (K562DRneo) [25] or pimEJ5GFP was grown in the same medium as K562 but under constant selection with 350 

HDACi decrease NHEJ repair efficiency

Several lines of evidence suggest that inhibition of HDACs decrease the expression of DSB repair proteins involved in NHEJ (i.e. Ku70, Ku80, DNA-PKcs) [14], [20], [39]. These effects contribute to decreased NHEJ but the exact mechanism is not clear [40]. We first tested whether pan-HDACi TSA decreases total cellular NHEJ activity in well-established leukemia cell lines. Using the previously published LacZα reactivation NHEJ repair assay [25], we found that indeed treatment of K562 (Fig. 1A) and

Discussion

Acetylation and deacetylation is a dynamic process that can have profound effects on the outcome of DSB repair. Several lines of evidence demonstrate that HDACi lead to decreased NHEJ activity [40], [31], but the mechanisms by which these inhibitors promote their effects on DSB repair remain unclear. We show here that pan-HDACi (TSA) inhibits the deacetylation of not only Ku70 or Ku80, but also PARP1, which functions in multiple DNA repair pathways including SSB and DSB repair. While HDACi

Authorship

Contributions: C.R., P.N. designed, performed experiments and wrote the paper. F.R. designed experiments and wrote the paper, N.P., A.A. performed experiments, I.G. furnished patient materials and advice in patient-based studies, P.C. and M.D.J. synthesized and furnished p300 small molecule inhibitors.

Conflicts of interest

The authors declare no conflict of interests.

Acknowledgements

We thank Hsing-Chen Tsai (JHU) for her help in designing the ChIP experiments, Stephen Baylin (JHU) and Alan Tomkinson (University of New Mexico) for their helpful comments. This work was supported by the National Institutes of Health (NIH) grants #E1905 (F.V.R.), NIH and FAMRI (P.C. and M.D.J.), NIH grant (CA186974) (F.V.R). The clinical study was supported in part by Cooperative Agreement from the National Cancer Institute (UO1CA69854) (I.G.).

References (55)

  • G.L. Dianov et al.

    Co-ordination of DNA single strand break repair

    DNA Repair

    (2007)
  • B. Paap et al.

    Human abasic endonuclease action on multilesion abasic clusters: implications for radiation-induced biological damage

    Nucleic Acids Res.

    (2008)
  • L.K. Jones et al.

    Chromatin modification, leukaemia and implications for therapy

    Br. J. Haematol.

    (2002)
  • J.E. Bolden et al.

    Anticancer activities of histone deacetylase inhibitors

    Nat. Rev. Drug Discov.

    (2006)
  • A. Kuendgen et al.

    Current status of epigenetic treatment in myelodysplastic syndromes

    Ann. Hematol.

    (2008)
  • J. Wang

    Caspase-mediated cleavage of ATM during cisplatin-induced tubular cell apoptosis: inactivation of its kinase activity toward p53

    Am. J. Physiol. Renal Physiol.

    (2006)
  • H.L. Alakomi et al.

    Weakening effect of cell permeabilizers on gram-negative bacteria causing biodeterioration

    Appl. Environ. Microbiol.

    (2006)
  • S. Furtak et al.

    Evaluation of the effect of methadone maintenance treatment program on the frequency of DNA lesions revealed as micronuclei and sister chromatid exchanges

    Przegl. Lek.

    (2005)
  • G. Paajarvi et al.

    HMG-coA reductase inhibitors, statins, induce phosphorylation of Mdm2 and attenuate the p53 response to DNA damage

    FASEB J.

    (2005)
  • A. Insinga

    Inhibitors of histone deacetylases induce tumor-selective apoptosis through activation of the death receptor pathway

    Nature

    (2005)
  • A. Nebbioso

    Tumor-selective action of HDAC inhibitors involves TRAIL induction in acute myeloid leukemia cells

    Nat. Med.

    (2005)
  • A.A. Ruefli

    The histone deacetylase inhibitor and chemotherapeutic agent suberoylanilide hydroxamic acid (SAHA) induces a cell-death pathway characterized by cleavage of Bid and production of reactive oxygen species

    Proc. Natl. Acad. Sci. U. S. A.

    (2001)
  • A. Munshi

    Histone deacetylase inhibitors radiosensitize human melanoma cells by suppressing DNA repair activity

    Clin. Cancer Res.

    (2005)
  • T.J. Gaymes

    Histone deacetylase inhibitors (HDI) cause DNA damage in leukemia cells: a mechanism for leukemia-specific HDI-dependent apoptosis?

    Mol. Cancer Res.

    (2006)
  • C.J. Bakkenist et al.

    DNA damage activates ATM through intermolecular autophosphorylation and dimer dissociation

    Nature

    (2003)
  • K. Camphausen

    Enhanced radiation-induced cell killing and prolongation of gammaH2AX foci expression by the histone deacetylase inhibitor MS-275

    Cancer Res.

    (2004)
  • T.C. Karagiannis et al.

    The histone deacetylase inhibitor, Trichostatin A, enhances radiation sensitivity and accumulation of gammaH2A.X

    Cancer Biol. Ther.

    (2005)
  • Cited by (57)

    • Epigenetic therapy and DNA damage response

      2022, Epigenetics and DNA Damage
    • Sp1-dependent recruitment of the histone acetylase p300 to DSBs facilitates chromatin remodeling and recruitment of the NHEJ repair factor Ku70

      2021, DNA Repair
      Citation Excerpt :

      p300 is shown to acetylate both Ku70/80 and PARP1 [57,58], and therefore inhibition of HAT proteins should result in decreased acetylation of Ku70/80 and PARP1. Whereas Ku70/80 acetylation decreases slightly with a p300 inhibitor, acetylation of PARP1 decreases significantly [59]. Additionally, p300 has been shown to transcriptionally activate both BRCA1 and Rad51, and inhibition of p300 reduces H3 and H4 acetylation and inhibits binding of the transcription factor E2F1 to these promoters [34].

    View all citing articles on Scopus
    1

    These authors contributed equally to the work.

    View full text