Research Article
Cell cycle-dependent DNA damage signaling induced by ICRF-193 involves ATM, ATR, CHK2, and BRCA1

https://doi.org/10.1016/j.yexcr.2006.02.029Get rights and content

Abstract

Topoisomerase II is essential for cell proliferation and survival and has been a target of various anticancer drugs. ICRF-193 has long been used as a catalytic inhibitor to study the function of topoisomerase II. Here, we show that ICRF-193 treatment induces DNA damage signaling. Treatment with ICRF-193 induced G2 arrest and DNA damage signaling involving γ-H2AX foci formation and CHK2 phosphorylation. DNA damage by ICRF-193 was further demonstrated by formation of the nuclear foci of 53BP1, NBS1, BRCA1, MDC1, and FANCD2 and increased comet tail moment. The DNA damage signaling induced by ICRF-193 was mediated by ATM and ATR and was restricted to cells in specific cell cycle stages such as S, G2, and mitosis including late and early G1 phases. Downstream signaling of ATM and ATR involved the phosphorylation of CHK2 and BRCA1. Altogether, our results demonstrate that ICRF-193 induces DNA damage signaling in a cell cycle-dependent manner and suggest that topoisomerase II might be essential for the progression of the cell cycle at several stages including DNA decondensation.

Introduction

Topoisomerases relax the superhelical tension of DNA. Type II topoisomerases are able to break and rejoin the two strands that make up duplex DNA. The activity of topoisomerase II (topo II) is essential for proliferating cell survival and participates in virtually all processes involving double-stranded DNA including replication, transcription, recombination, chromosome condensation, and the decatenation of sister chromatids prior to the anaphase of mitosis [1]. In cancer chemotherapy, topo II is one of the major targets for a variety of anticancer drugs. According to their mechanism of action, these drugs have been classified into two groups. One class of drugs termed topo II poisons, including anthracyclines (adriamycin/doxorubicin and daunorubicin), epipodophyllotoxins (etoposide/VP-16 and teniposide/VM-26), anthracenedione (mitoxantrone), isoflavonoid (genistein), and aminoacridines (amsacrine/m-AMSA), stabilizes the protein-linked DNA intermediate “cleavable complex” and produces DNA double-strand breaks (DSB) through this complex [2]. Topo II poisons are much more cytotoxic than the other class of drugs, topo II catalytic inhibitors. Topo II catalytic inhibitors that do not stabilize the cleavable complex inhibit topo II by locking topo II in a “closed clamp,” thus preventing strand passage [3]. Bis-dioxopiperazines (ICRF-154, ICRF-187, ICRF-193, etc.), fostriecin, aclarubicin, suramin, novobiocin, and merbarone all belong to this class of drugs.

DNA damage induced by ionizing radiation (IR), ultraviolet radiation (UV), or abnormal structures such as stalled replication forks generally results in the rapid activation of DNA damage signaling pathways, cell cycle arrest, and DNA repair, with the overall purpose of maintaining genome stability. In vertebrates, ataxia-telangiectasia mutated (ATM) and ATM- and Rad3-related (ATR), members of the phosphatidylinositol 3-kinase-related protein family, are critical checkpoint regulators which work upstream of the DNA damage response pathway [4]. In humans, ATM is mutated in the autosomal disorder, ataxia-telangiectasia (A-T) syndrome. These patients display an increased rate of chromosomal recombination and are defective in IR-induced G1/S, S phase, and G2/M checkpoints. ATM seems to be more specifically involved in responses to DSB, whereas one human disease, ATR-Seckel syndrome, has been reported to have ATR deficiency [5], and in mice, ATR disruption leads to early embryonic lethality [6]. ATR has been shown to regulate responses to a broad range of damage, including stalled replication forks, UV-induced photodimers, nucleotide depletion, polymerase arrest, interstrand crosslinks, and DSB [7], [8]. The checkpoint functions of ATR and ATM are mediated in part by a pair of checkpoint effector kinases termed CHK1 and CHK2 [9], [10]. Histone H2AX, 53BP1, BRCA1, MDC1, FANCD2, and NBS1 are all targets for ATM- or ATR-mediated phosphorylation [11], [12], [13], [14], [15], [16], [17], [18]. These molecules participate in the transmission of DNA damage signals to downstream molecules such as CHK1 and CHK2 and colocalize to foci containing the site of damaged DNA. These foci are presumed to be checkpoint/repair factories. Whereas the phosphorylation of CHK1 by ATR is induced by IR, UV, stalled replication forks, and upon activation of the mismatch repair system by 6-thioguanine or methylating agents [9], [19], CHK2 is phosphorylated by ATM in response to IR, stalled replication forks, and activation of the mismatch repair system by 6-thioguanine or methylating agents [4], [19], [20]. The topo II poisons, doxorubicin, genistein, and etoposide, induce DSB in which the signal is transduced through CHK2 in an ATM-dependent manner [21], [22].

ICRF-193 has been extensively analyzed as a topo II catalytic inhibitor to study the function of topo II [23]. ICRF-193-treated cells delay G2/M transition as well as the progression from metaphase to anaphase in mammalian cells [24]. The nature of this G2 delay by ICRF-193 treatment has been proposed as a “decatenation checkpoint,” in which cells monitor chromatid catenation status after DNA replication and inhibit progression into mitosis until the chromatids are correctly decatenated by topo II [25]. Activation of the decatenation G2 checkpoint relies on ATR activity and the subsequent nuclear exclusion of cyclin B1. However, several recent observations suggest that ICRF-193 may induce DNA damage in vivo and in vitro, although the extent of DNA damage is weak as compared to that induced by topo II poisons [26], [27], [28], [29], [30], [31].

Although several reports suggest that ICRF-193 can induce DNA damage, this issue is still controversial. Moreover, the mechanism by which ICRF-193 induces DNA damage has not been studied extensively. We initiated this study with the aim of understanding the mechanism of G2 arrest by ICRF-193 treatment. Here, we show that ICRF-193 induced DNA damage resulting in G2 arrest and that DNA damage signaling by ICRF-193 involved molecules reminiscent of those participating in DSB by IR. In addition, cell cycle-dependent DNA damage induced by ICRF-193 treatment demonstrated that topo II is essential for the progression of the cell cycle at several stages, including S, G2, and mitosis [1]. Lastly, for the first time in mammalian cells, we provide evidence that topo II is required during late mitosis and the early G1 phase, presumably for chromosome decondensation.

Section snippets

Cell lines and culture conditions

HeLa cells (purchased from ATCC) were maintained in DMEM supplemented with 10% fetal calf serum. GM16666 and GM16667 cells (purchased from Coriell Cell Repositories) were maintained in DMEM supplemented with 10% fetal calf serum, 2 mM of l-glutamine, and 100 μg/ml of hygromycin B. The ATM-deficient (A-T) fibroblasts immortalized with hTERT and normal human fibroblasts immortalized with hTERT (generously provided by Dr. Pandita at Washington University School of Medicine, St. Louis, MO) were

ICRF-193 induces DNA damage and nuclear foci formation of γ-H2AX, NBS1, BRCA1, 53BP1, MDC1, and FANCD2

To test whether ICRF-193 treatment induces DNA damage, the nuclear foci formation of proteins including γ-H2AX, NBS1, BRCA1, 53BP1, MDC1, and FANCD2 was examined in HeLa cells. Phosphorylation of histone H2AX is among the earliest responses to DNA damage [33], [34]. We found that 6 h of treatment with 10 μM ICRF-193 induced the formation of γ-H2AX, NBS1, BRCA1, 53BP1, MDC1, and FANCD2 nuclear foci (Fig. 1A). Induction of γ-H2AX foci was observed after ICRF-193 treatment, but the kinetics of the

Discussion

To explore the function of topo II, various inhibitors have been used, including poisons and catalytic inhibitors. Whereas topo II poisons induce DNA damage by forming a “cleavable complex,” catalytic inhibitors of topo II are generally regarded as not inducing DNA damage and just inhibiting the catalytic activity of the enzyme [3]. For these reasons, catalytic inhibitors of topo II are preferentially used to study the function of topo II. Whereas several recent observations suggest that

Acknowledgments

This work was supported in part by the National Institutes of Health Grants K18HL74106 (to H.A.), by CA 96805 (to H.A.), and by the Department of Defense grants W81XWH-04-1-0721 and W81XWH-04-1-0711. We thank W.K. Kaufmann for the generous gift of ATR-inducible GM847 cells, T.K. Pandita for the hTERT-immortalized normal fibroblasts and A-T fibroblasts, S.J. Elledge for polyclonal antibodies against MDC1, and T.D. Halazonetis for monoclonal antibody against 53BP1. We also thank R. Hauser and A.

References (53)

  • E.P. Rogakou et al.

    DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139

    J. Biol. Chem.

    (1998)
  • N. Pastor et al.

    High yield of endoreduplication induced by ICRF-193: a topoisomerase II catalytic inhibitor

    Mutat. Res.

    (2002)
  • N. Akimitsu et al.

    Induction of apoptosis by depletion of DNA topoisomerase IIα in mammalian cells

    Biochem. Biophys. Res. Commun.

    (2003)
  • R. Scully et al.

    Association of BRCA1 with Rad51 in mitotic and meiotic cells

    Cell

    (1997)
  • D.A. Skoufias et al.

    Inhibition of DNA decatenation, but not DNA damage, arrests cells at metaphase

    Mol. Cell

    (2004)
  • S.J. Haggarty et al.

    Small molecule modulation of the human chromatid decatenation checkpoint

    Chem. Biol.

    (2003)
  • N. Adachi et al.

    Genetic evidence for involvement of two distinct nonhomologous end-joining pathways in repair of topoisomerase II-mediated DNA damage

    Biochem. Biophys. Res. Commun.

    (2004)
  • N. Adachi et al.

    Hypersensitivity of nonhomologous DNA end-joining mutants to VP-16 and ICRF-193: implications for the repair of topoisomerase II-mediated DNA damage

    J. Biol. Chem.

    (2003)
  • M. Kobayashi et al.

    Decreased topoisomerase IIα expression confers increased resistance to ICRF-193 as well as VP-16 in mouse embryonic stem cells

    Cancer Lett.

    (2001)
  • J.C. Wang

    Cellular roles of DNA topoisomerases: a molecular perspective

    Nat. Rev., Mol. Cell Biol.

    (2002)
  • Y. Shiloh

    ATM and related protein kinases: safeguarding genome integrity

    Nat. Rev., Cancer

    (2003)
  • G.K. Alderton et al.

    Seckel syndrome exhibits cellular features demonstrating defects in the ATR-signalling pathway

    Hum. Mol. Genet.

    (2004)
  • E.J. Brown et al.

    ATR disruption leads to chromosomal fragmentation and early embryonic lethality

    Genes Dev.

    (2000)
  • E.J. Brown et al.

    Essential and dispensable roles of ATR in cell cycle arrest and genome maintenance

    Genes Dev.

    (2003)
  • R.S. Tibbetts et al.

    Functional interactions between BRCA1 and the checkpoint kinase ATR during genotoxic stress

    Genes Dev.

    (2000)
  • Q. Liu et al.

    Chk1 is an essential kinase that is regulated by Atr and required for the G(2)/M DNA damage checkpoint

    Genes Dev.

    (2000)
  • Cited by (40)

    • Evaluation of the genotoxic potential of apoptosis inducers with the γH2AX assay in human cells

      2020, Mutation Research - Genetic Toxicology and Environmental Mutagenesis
    • Mechanism of the inhibition of leukemia cell growth and induction of apoptosis through the activation of ATR and PTEN by the topoisomerase inhibitor 3EZ, 20Ac-ingenol

      2015, Leukemia Research
      Citation Excerpt :

      In contrast, transcription independent p53 apoptosis is mediated by interactions of mitochondrial p53 with Bcl-2 [46]. Although the class II catalytic inhibitors, ICRF-193 and merbarone, and the catalytic inhibitor of topoisomerase I, betulinic acid, are not known to induce apoptosis via Bcl-2 genes [20,22,32,39], the class I poison inhibitor of topoisomerase I, CPT and the catalytic inhibitor, β-lapachone, stimulates this mitochondrial apoptosis pathway [33,42,43]. In the present experiment, p53 upregulation was sustained for 24 h of exposure to 3EZ, 20Ac-ingenol, and Bax expression was elevated, Bcl-2 protein was downregulated, and caspase 3 was activated at this time point.

    • Design and synthesis of 2-phenylnaphthalenoids as inhibitors of DNA topoisomeraseIIα and antitumor agents

      2014, European Journal of Medicinal Chemistry
      Citation Excerpt :

      One class of TopoII inhibitors, namely the TopoII poisons, such as doxorubicin and etoposide (VP16), are believed to stabilize the drug-Topo-DNA cleavable complex and enhance DNA double strand breaks [6,7]. Another class of TopoII-targeted agents, the TopoII catalytic inhibitors, such as bis(2,6-dioxopiperazine) derivatives ICRF-193, do not stabilize the cleavable complex but inhibit the catalytic activity of the TopoII [8–10]. Among the TopoII inhibitors in clinical uses, the TopoII poisons are much more popular than the TopoII catalytic inhibitors [10,11].

    • Alternariol induces abnormal nuclear morphology and cell cycle arrest in murine RAW 264.7 macrophages

      2013, Toxicology Letters
      Citation Excerpt :

      The presence of a decatenation checkpoint that is independent of DNA damage has been supported by a variety of studies (Damelin et al., 2005; Nakagawa et al., 2004). The concept is however still controversial as the topo II inhibitor ICRF-193 has been shown to activate DNA damage signaling in some cancer cell lines (Park and Avraham, 2006). Mitosis can be subdivided into karyokinesis (prophase, prometaphase, metaphase, anaphase, telophase) and cytokinesis.

    View all citing articles on Scopus
    View full text