Elsevier

DNA Repair

Volume 5, Issue 8, 13 August 2006, Pages 904-913
DNA Repair

Impaired elimination of DNA double-strand break-containing lymphocytes in ataxia telangiectasia and Nijmegen breakage syndrome

https://doi.org/10.1016/j.dnarep.2006.05.002Get rights and content

Abstract

The repair of DNA double-strand breaks is critical for genome integrity and tumor suppression. Here we show that following treatment with the DNA-intercalating agent actinomycin D (ActD), normal quiescent T cells accumulate double-strand breaks and die, whereas T cells from ataxia telangiectasia (AT) and Nijmegen breakage syndrome (NBS) patients are resistant to this death pathway despite a comparable amount of DNA damage. We demonstrate that the ActD-induced death pathway in quiescent T lymphocytes follows DNA damage and H2AX phosphorylation, is ATM- and NBS1-dependent and due to p53-mediated cellular apoptosis. In response to genotoxic 2-Gy γ-irradiation, on the other hand, quiescent T cells from normal donors survive following complete resolution of the damage thus induced. T cells from AT and NBS patients also survive, but retain foci of phosphorylated H2AX due to a subtle double-strand break (DSB) repair defect. A common consequence of these two genetic defects in the DSB response is the apparent tolerance of cells containing DNA breaks. We suggest that this tolerance makes a major contribution to the oncogenic risk of patients with chromosome instability syndromes.

Introduction

DSB repair is critical for genome integrity and tumor suppression. DSBs are produced either by exogenous agents or by endogenous events, such as collapsed replication forks and gene rearrangements during lymphocyte development. Antigen receptor gene recombination is initiated by the recombinase-activating gene 1/2 (RAG1/2) endonuclease during the G0/G1 phase of the cell cycle [1]. This generates DSBs adjacent to the V, D and J segments [2] that are subsequently compiled as complete V(D)J coding exons by nonhomologous end joining [3].

V(D)J recombination is now known to be active in precursor bone marrow and thymic cells, and in a fraction of mature B and T cells that undergo receptor revision in the periphery [4], [5]. We and others have observed functional RAG gene products in mature T cells featuring defective TCR expression [6], [7]. The frequency of this T cell subset is increased more than 10-fold in patients with AT and NBS [8], [9], two autosomal recessive genomic instability syndromes characterized by immunodeficiency, hypersensitivity to ionizing radiation (IR) and DSB-inducing agents, defective DNA repair, and an increased risk of lymphoid malignancies [10], [11], [12], [13]. The role of V(D)J recombination in the onset of tumorigenic translocations is well established [14], [15], [16], [17], and a similar role has been suggested for secondary V(D)J rearrangements [18]. Several mechanisms have evolved in normal cells to counter the dangers of V(D)J recombination, i.e. the specific targeting of DNA DSBs to the intended sites, their efficient rejoining and the activation by unresolved DSBs of a general DNA damage-response [19]. Alterations of any of these processes, together with increased RAG activity, in lymphocytes defective for the ATM or NBS1 proteins would provide an explanation of their increased risk of leukemia/lymphoma [20].

We here assessed the response of normal, AT and NBS T cells kept in the G0/G1 phase to two genotoxic agents: ActD and IR. ActD is an anti-tumor antibiotic used in many chemotherapeutical protocols that intercalates into DNA and thus inhibits transcription and induces damage [21], [22]. We show that ActD produces DSBs in normal quiescent T lymphocytes and targets them to apoptosis, whereas T cells from AT and NBS patients resist this apoptosis and survive. Following IR (2-Gy) quiescent T cells from normal donors resolve virtually all damage within 72 h and survive. T cells from AT and NBS patients also survive, despite the maintenance of γH2AX foci.

Inefficient elimination of DSB-containing cells may be supposed to make a major contribution to the oncogenic risk of patients with chromosome instability syndromes.

Section snippets

Cell isolation, cell culture and induction of DNA damage

Peripheral blood from four healthy donors, six AT and four NBS patients was collected after signed informed consent. AT patients were either homozygous or compound heterozygous for truncating mutations; NBS patients were homozygous for the common 657del5 mutation. PBMC were isolated as described [7] and either used within 2 days or frozen and used immediately after thawing. In all cases, >95% of cells were in G0/G1. T cell lines were generated as described [7]. All experiments were conducted on

Resting T lymphocytes from AT and NBS patients are resistant to ActD-induced apoptosis

To define the sensitivity to ActD of quiescent circulating lymphocytes, freshly separated PBMC from four healthy donors were treated with 0.5 μg/ml ActD and analyzed by flow cytometry with propidium iodide (PI) staining (Fig. 1a). Mature lymphocytes were susceptible to ActD-induced death (mean 19% viable cells at 72 h). In contrast, mature lymphocytes from three AT and three NBS patients were resistant. Their viability at 72 h exceeded 60%: AT versus control, p = 0.0003; NBS versus control, p = 0.0001

Discussion

Here we show that quiescent AT and NBS T lymphocytes are resistant to an apoptotic death pathway induced in normal T cells by ActD and display a subtle, but defined DSB repair defect following IR. A common consequence of these two different alterations in the DSB response is apparent tolerance of cells containing DSB.

The ability of AT and NBS T cells to survive DNA damage induced by intercalating agents is only shared with immature thymocytes [40 and our unpublished results], where it reflects

Acknowledgements

We thank Drs. G. Forni, University of Turin, Italy, and G. Del Sal, University of Trieste, Italy, for their critical reading of the MS and comments. We gratefully acknowledge the contribution of Drs. A. Brusco, University of Turin, Italy, and A. Marchi, University of Pavia, Italy, for providing biological samples from AT patients. We thank Dr. A. Lanzavecchia, IRB, Bellinzona, Switzerland for the rIL2-producing myeloma system and Dr. M. Moretti, University of Perugia, Italy for help with the

References (60)

  • J.J. Cohen

    Apoptosis

    Immunol. Today

    (1993)
  • E.P. Rogakou et al.

    Initiation of DNA fragmentation during apoptosis induces phosphorylation of H2AX histone at serine 139

    J. Biol. Chem.

    (2000)
  • A. Blasina et al.

    Caffeine inhibits the checkpoint kinase ATM

    Curr. Biol.

    (1999)
  • E. Riballo et al.

    A pathway of double-strand break rejoining dependent upon ATM, Artemis, and proteins locating to gamma-H2AX foci

    Mol. Cell.

    (2004)
  • I.I. Kruman et al.

    Cell cycle activation linked to neuronal cell death initiated by DNA damage

    Neuron

    (2004)
  • C. Barlow et al.

    Atm-deficient mice: a paradigm of ataxia telangiectasia

    Cell

    (1996)
  • K.M. Cerosaletti et al.

    Nibrin forkhead-associated domain and breast cancer C-terminal domain are both required for nuclear focus formation and phosphorylation

    J. Biol. Chem.

    (2003)
  • K. Andreau et al.

    Preapoptotic chromatin condensation upstream of the mitochondrial checkpoint

    J. Biol. Chem.

    (2004)
  • M. Lobrich et al.

    Harmonising the response to DSBs: a new string in the ATM bow

    DNA Repair

    (2005)
  • D.W. Meek

    The p53 response to DNA damage

    DNA Repair

    (2004)
  • S. Desiderio et al.

    The cell cycle and V(D)J recombination

    Curr. Top. Microbiol. Immunol.

    (1996)
  • S.D. Fugmann et al.

    The RAG proteins and V(D)J recombination: complexes, ends, and transposition

    Annu. Rev. Immunol.

    (2000)
  • C.H. Bassing et al.

    Increased ionizing radiation sensitivity and genomic instability in the absence of histone H2AX

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

    (2002)
  • E. Lantelme et al.

    Cutting edge: recombinase-activating gene expression and V(D)J recombination in CD4+CD3low mature T lymphocytes

    J. Immunol.

    (2000)
  • E. Lantelme et al.

    Increased frequency of RAG-expressing, CD4(+)CD3(low) peripheral T lymphocytes in patients with defective responses to DNA damage

    Eur. J. Immunol.

    (2000)
  • Y. Shiloh

    ATM and related protein kinases: safeguarding genome integrity

    Nat. Rev. Cancer

    (2003)
  • S.G. Becker-Catania et al.

    Ataxia-telangiectasia

    Adv. Exp. Med. Biol.

    (2001)
  • Y. Shiloh

    Ataxia-telangiectasia and the Nijmegen breakage syndrome: related disorders but genes apart

    Annu. Rev. Genet.

    (1997)
  • F.G. Haluska et al.

    The t(8; 14) chromosomal translocation occurring in B-cell malignancies results from mistakes in V–D–J joining

    Nature

    (1986)
  • T.H. Rabbitts

    Chromosomal translocations in human cancer

    Nature

    (1994)
  • Cited by (0)

    1

    These authors contributed equally to this work.

    View full text