A novel interplay between the Fanconi anemia core complex and ATR-ATRIP kinase during DNA cross-link repair

When DNA replication is stalled at sites of DNA damage, a cascade of responses is activated in the cell to halt cell cycle progression and promote DNA repair. A pathway initiated by the kinase Ataxia teleangiectasia and Rad3 related (ATR) and its partner ATR interacting protein (ATRIP) plays an important role in this response. The Fanconi anemia (FA) pathway is also activated following genomic stress, and defects in this pathway cause a cancer-prone hematologic disorder in humans. Little is known about how these two pathways are coordinated. We report here that following cellular exposure to DNA cross-linking damage, the FA core complex enhances binding and localization of ATRIP within damaged chromatin. In cells lacking the core complex, ATR-mediated phosphorylation of two functional response targets, ATRIP and FANCI, is defective. We also provide evidence that the canonical ATR activation pathway involving RAD17 and TOPBP1 is largely dispensable for the FA pathway activation. Indeed DT40 mutant cells lacking both RAD17 and FANCD2 were synergistically more sensitive to cisplatin compared with either single mutant. Collectively, these data reveal new aspects of the interplay between regulation of ATR-ATRIP kinase and activation of the FA pathway.

[1]  L. Zou,et al.  ATR: a master conductor of cellular responses to DNA replication stress. , 2011, Trends in biochemical sciences.

[2]  A. D’Andrea,et al.  Expanded roles of the Fanconi anemia pathway in preserving genomic stability. , 2010, Genes & development.

[3]  Stephen J. Elledge,et al.  Sensing DNA Damage Through ATRIP Recognition of RPA-ssDNA Complexes , 2003, Science.

[4]  A. Auerbach,et al.  Mutations of the SLX4 gene in Fanconi anemia , 2011, Nature Genetics.

[5]  Y. Maehara,et al.  ATR-ATRIP kinase complex triggers activation of the Fanconi anemia DNA repair pathway. , 2012, Cancer research.

[6]  H. Arakawa,et al.  Fanconi Anemia Protein FANCD2 Promotes Immunoglobulin Gene Conversion and DNA Repair through a Mechanism Related to Homologous Recombination , 2005, Molecular and Cellular Biology.

[7]  K. Yoshioka,et al.  Disruption of ATM in p53-null cells causes multiple functional abnormalities in cellular response to ionizing radiation , 1999, Oncogene.

[8]  H. Kurumizaka,et al.  KIAA1018/FAN1 nuclease protects cells against genomic instability induced by interstrand cross-linking agents , 2010, Proceedings of the National Academy of Sciences.

[9]  A. Matsuura,et al.  Dimerization of the ATRIP protein through the coiled-coil motif and its implication to the maintenance of stalled replication forks. , 2005, Molecular biology of the cell.

[10]  H. Kurumizaka,et al.  Involvement of SLX4 in interstrand cross-link repair is regulated by the Fanconi anemia pathway , 2011, Proceedings of the National Academy of Sciences.

[11]  H. Kimura,et al.  Histone chaperone activity of Fanconi anemia proteins, FANCD2 and FANCI, is required for DNA crosslink repair , 2012, The EMBO journal.

[12]  Franca Fraternali,et al.  Mutation of the RAD51C gene in a Fanconi anemia–like disorder , 2010, Nature Genetics.

[13]  K. Cimprich,et al.  ATR: an essential regulator of genome integrity , 2008, Nature Reviews Molecular Cell Biology.

[14]  M. Kimura,et al.  Amino‐terminal domain of ATRIP contributes to intranuclear relocation of the ATR–ATRIP complex following DNA damage , 2004, FEBS letters.

[15]  Anindya Dutta,et al.  UBE2T, the Fanconi Anemia Core Complex, and FANCD2 Are Recruited Independently to Chromatin: a Basis for the Regulation of FANCD2 Monoubiquitination , 2007, Molecular and Cellular Biology.

[16]  F. Rosselli,et al.  BLM and the FANC proteins collaborate in a common pathway in response to stalled replication forks , 2004, The EMBO journal.

[17]  D. Schindler,et al.  SLX4, a coordinator of structure-specific endonucleases, is mutated in a new Fanconi anemia subtype , 2011, Nature Genetics.

[18]  S. West,et al.  Identification of FAAP24, a Fanconi anemia core complex protein that interacts with FANCM. , 2007, Molecular cell.

[19]  David A. Williams,et al.  Impaired FANCD2 monoubiquitination and hypersensitivity to camptothecin uniquely characterize Fanconi anemia complementation group M. , 2009, Blood.

[20]  S. Elledge,et al.  FANCI phosphorylation functions as a molecular switch to turn on the Fanconi anemia pathway , 2008, Nature Structural &Molecular Biology.

[21]  J. Gautier,et al.  Checkpoint signaling from a single DNA interstrand crosslink. , 2009, Molecular cell.

[22]  Miki Suzuki,et al.  Dynamics of human replication factors in the elongation phase of DNA replication , 2007, Nucleic acids research.

[23]  E. Levy-Lahad,et al.  Fanconi anemia and breast cancer susceptibility meet again , 2010, Nature Genetics.

[24]  A. D’Andrea,et al.  A new nuclease member of the FAN club , 2010, Nature Structural &Molecular Biology.

[25]  J. Gautier,et al.  Fanconi anemia proteins stabilize replication forks. , 2008, DNA repair.

[26]  R. Schwab,et al.  ATR activation and replication fork restart are defective in FANCM‐deficient cells , 2010, The EMBO journal.

[27]  A. Gurtan,et al.  Cell cycle-dependent chromatin loading of the Fanconi anemia core complex by FANCM/FAAP24. , 2008, Blood.

[28]  H. Kurumizaka,et al.  DNA robustly stimulates FANCD2 monoubiquitylation in the complex with FANCI , 2012, Nucleic acids research.

[29]  D. Cortez,et al.  Activation of ATR and related PIKKs , 2008, Cell cycle.

[30]  H. Joenje,et al.  Fancm-deficient mice reveal unique features of Fanconi anemia complementation group M. , 2009, Human molecular genetics.

[31]  Hans Joenje,et al.  FANCE: the link between Fanconi anaemia complex assembly and activity , 2002, The EMBO journal.

[32]  D. Cortez,et al.  ATRIP Oligomerization Is Required for ATR-dependent Checkpoint Signaling* , 2005, Journal of Biological Chemistry.

[33]  Linda Z. Shi,et al.  CtIP Is Required to Initiate Replication-Dependent Interstrand Crosslink Repair , 2012, PLoS genetics.

[34]  A. Sobeck,et al.  Fanconi anemia proteins FANCD2 and FANCI exhibit different DNA damage responses during S-phase , 2012, Nucleic acids research.

[35]  J. Sale,et al.  Deubiquitination of FANCD2 Is Required for DNA Crosslink Repair , 2007, Molecular cell.

[36]  F. Rosselli,et al.  The DNA crosslink‐induced S‐phase checkpoint depends on ATR–CHK1 and ATR–NBS1–FANCD2 pathways , 2004, The EMBO journal.

[37]  S. Elledge,et al.  Identification of the FANCI Protein, a Monoubiquitinated FANCD2 Paralog Required for DNA Repair , 2007, Cell.

[38]  Jun Qin,et al.  Involvement of the TIP60 Histone Acetylase Complex in DNA Repair and Apoptosis , 2000, Cell.

[39]  J. Glover,et al.  ATR autophosphorylation as a molecular switch for checkpoint activation. , 2011, Molecular cell.

[40]  H. Kitao,et al.  A FancD2-monoubiquitin fusion reveals hidden functions of Fanconi anemia core complex in DNA repair. , 2005, Molecular cell.

[41]  Hiroshi Arakawa,et al.  Functional relationships of FANCC to homologous recombination, translesion synthesis, and BLM , 2005, The EMBO journal.

[42]  W. Lambert,et al.  Damage-resistant DNA synthesis in Fanconi anemia cells treated with a DNA cross-linking agent. , 2000, Experimental cell research.

[43]  M. Wold,et al.  Recombinant replication protein A: expression, complex formation, and functional characterization. , 1994, The Journal of biological chemistry.

[44]  D. Papadopoulo,et al.  Arrest of S-phase progression is impaired in Fanconi anemia cells. , 2000, Experimental cell research.

[45]  A. D’Andrea,et al.  The FANCM/FAAP24 complex is required for the DNA interstrand crosslink-induced checkpoint response. , 2010, Molecular cell.

[46]  J. Pereira-Leal,et al.  The vertebrate Hef ortholog is a component of the Fanconi anemia tumor-suppressor pathway , 2005, Nature Structural &Molecular Biology.

[47]  J. Myers,et al.  ATRIP binding to replication protein A-single-stranded DNA promotes ATR-ATRIP localization but is dispensable for Chk1 phosphorylation. , 2005, Molecular biology of the cell.

[48]  David J Adams,et al.  Disruption of mouse Slx4, a regulator of structure-specific nucleases, phenocopies Fanconi Anemia , 2011, Nature Genetics.

[49]  A. D’Andrea,et al.  ATR couples FANCD2 monoubiquitination to the DNA-damage response. , 2004, Genes & development.

[50]  K. J. Patel,et al.  The Walker B motif in avian FANCM is required to limit sister chromatid exchanges but is dispensable for DNA crosslink repair , 2009, Nucleic acids research.

[51]  O. Matsui,et al.  Critical role for chicken Rad17 and Rad9 in the cellular response to DNA damage and stalled DNA replication , 2004, Genes to cells : devoted to molecular & cellular mechanisms.

[52]  S. Elledge,et al.  How ATR turns on: TopBP1 goes on ATRIP with ATR. , 2008, Genes & development.

[53]  A. D’Andrea,et al.  Regulation of DNA cross-link repair by the Fanconi anemia/BRCA pathway. , 2012, Genes & development.

[54]  Weidong Wang Emergence of a DNA-damage response network consisting of Fanconi anaemia and BRCA proteins , 2007, Nature Reviews Genetics.

[55]  L. Zou,et al.  ATRIP associates with replication protein A-coated ssDNA through multiple interactions. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[56]  H. Kitao,et al.  A requirement of FancL and FancD2 monoubiquitination in DNA repair , 2007, Genes to cells : devoted to molecular & cellular mechanisms.

[57]  W. Chazin,et al.  Function of a Conserved Checkpoint Recruitment Domain in ATRIP Proteins , 2007, Molecular and Cellular Biology.

[58]  C. Vaziri,et al.  The Chk1-mediated S-phase Checkpoint Targets Initiation Factor Cdc45 via a Cdc25A/Cdk2-independent Mechanism* , 2006, Journal of Biological Chemistry.

[59]  S. Elledge,et al.  FANCM and FAAP24 function in ATR-mediated checkpoint signaling independently of the Fanconi anemia core complex. , 2008, Molecular cell.

[60]  Tom Walsh,et al.  Ten genes for inherited breast cancer. , 2007, Cancer cell.

[61]  A. Matsuura,et al.  ATR-dependent phosphorylation of ATRIP in response to genotoxic stress. , 2004, Biochemical and biophysical research communications.

[62]  Lei Li,et al.  FANCM and FAAP24 maintain genome stability via cooperative as well as unique functions. , 2013, Molecular cell.