Amendment history : Corrigendum ( March 2017 ) Biallelic inactivation of REV 7 is associated with Fanconi anemia

Fanconi anemia (FA) is a recessive genetic disease characterized by congenital abnormalities, chromosome instability, progressive bone marrow failure (BMF), and a strong predisposition to cancer. Twenty FA genes have been identified, and the FANC proteins they encode cooperate in a common pathway that regulates DNA crosslink repair and replication fork stability. We identified a child with severe BMF who harbored biallelic inactivating mutations of the translesion DNA synthesis (TLS) gene REV7 (also known as MAD2L2), which encodes the mutant REV7 protein REV7-V85E. Patient-derived cells demonstrated an extended FA phenotype, which included increased chromosome breaks and G2/M accumulation upon exposure to DNA crosslinking agents, gH2AX and 53BP1 foci accumulation, and enhanced p53/p21 activation relative to cells derived from healthy patients. Expression of WT REV7 restored normal cellular and functional phenotypes in the patient’s cells, and CRISPR/Cas9 inactivation of REV7 in a non-FA human cell line produced an FA phenotype. Finally, silencing Rev7 in primary hematopoietic cells impaired progenitor function, suggesting that the DNA repair defect underlies the development of BMF in FA. Taken together, our genetic and functional analyses identified REV7 as a previously undescribed FA gene, which we term FANCV. Brief Report Genetics Hematology

[1]  A. D’Andrea,et al.  The Fanconi anaemia pathway: new players and new functions , 2016, Nature Reviews Molecular Cell Biology.

[2]  J. Surrallés,et al.  Fanconi anemia: a model disease for studies on human genetics and advanced therapeutics. , 2015, Current opinion in genetics & development.

[3]  J. Sale REV7/MAD2L2: the multitasking maestro emerges as a barrier to recombination , 2015, The EMBO journal.

[4]  Peter Bouwman,et al.  REV7 counteracts DNA double-strand break resection and affects PARP inhibition , 2015, Nature.

[5]  David A. Williams,et al.  Exit from dormancy provokes DNA-damage-induced attrition in haematopoietic stem cells , 2015, Nature.

[6]  D. Durocher,et al.  MAD2L2 controls DNA repair at telomeres and DNA breaks by inhibiting 5′ end-resection , 2015, Nature.

[7]  R. Wood,et al.  REV7 is essential for DNA damage tolerance via two REV3L binding sites in mammalian DNA polymerase ζ , 2015, Nucleic acids research.

[8]  M. Gregory,et al.  Human Pol ζ purified with accessory subunits is active in translesion DNA synthesis and complements Pol η in cisplatin bypass , 2014, Proceedings of the National Academy of Sciences.

[9]  H. Yamanishi,et al.  A Missense Mutation in Rev7 Disrupts Formation of Polζ, Impairing Mouse Development and Repair of Genotoxic Agent-induced DNA Lesions* , 2013, The Journal of Biological Chemistry.

[10]  J. Sale,et al.  Sequestration of CDH1 by MAD2L2 prevents premature APC/C activation prior to anaphase onset , 2013, The Journal of cell biology.

[11]  M. Stojkovic,et al.  Brief Report: Human Pluripotent Stem Cell Models of Fanconi Anemia Deficiency Reveal an Important Role for Fanconi Anemia Proteins in Cellular Reprogramming and Survival of Hematopoietic Progenitors , 2013, Stem cells.

[12]  Y. Murakumo,et al.  The REV7 Subunit of DNA Polymerase ζ Is Essential for Primordial Germ Cell Maintenance in the Mouse* , 2013, The Journal of Biological Chemistry.

[13]  C. Canman,et al.  The roles of DNA polymerase ζ and the Y family DNA polymerases in promoting or preventing genome instability. , 2013, Mutation research.

[14]  Molly C. Kottemann,et al.  Fanconi anaemia and the repair of Watson and Crick DNA crosslinks , 2013, Nature.

[15]  K. J. Patel,et al.  Genotoxic consequences of endogenous aldehydes on mouse haematopoietic stem cell function , 2012, Nature.

[16]  J. Soulier,et al.  Bone marrow failure in Fanconi anemia is triggered by an exacerbated p53/p21 DNA damage response that impairs hematopoietic stem and progenitor cells. , 2012, Cell stem cell.

[17]  A. D’Andrea,et al.  Regulation of Rev1 by the Fanconi Anemia Core Complex , 2011, Nature Structural &Molecular Biology.

[18]  J. Soulier Fanconi anemia. , 2011, Hematology. American Society of Hematology. Education Program.

[19]  D. Pellman,et al.  Cytokinesis failure occurs in Fanconi anemia pathway-deficient murine and human bone marrow hematopoietic cells. , 2010, The Journal of clinical investigation.

[20]  M. Sivasubramaniam,et al.  Ku70 Corrupts DNA Repair in the Absence of the Fanconi Anemia Pathway , 2010, Science.

[21]  Z. Hořejší,et al.  Preventing nonhomologous end joining suppresses DNA repair defects of Fanconi anemia. , 2010, Molecular cell.

[22]  B. Alter,et al.  Pathophysiology and management of inherited bone marrow failure syndromes. , 2010, Blood reviews.

[23]  S. Elledge,et al.  The Fanconi Anemia Pathway Promotes Replication-Dependent DNA Interstrand Cross-Link Repair , 2009, Science.

[24]  A. D’Andrea,et al.  Mouse models of Fanconi anemia. , 2009, Mutation research.

[25]  F. Rosselli,et al.  The FANC pathway and BLM collaborate during mitosis to prevent micro-nucleation and chromosome abnormalities , 2009, Nature Cell Biology.

[26]  I. Hickson,et al.  Replication stress induces sister-chromatid bridging at fragile site loci in mitosis , 2009, Nature Cell Biology.

[27]  A. D’Andrea,et al.  The Fanconi anemia core complex is required for efficient point mutagenesis and Rev1 foci assembly. , 2008, DNA repair.

[28]  H. Saya,et al.  Multiple Roles of Vertebrate REV Genes in DNA Repair and Recombination , 2005, Molecular and Cellular Biology.

[29]  H. Broxmeyer,et al.  Fanconi anemia type C and p53 cooperate in apoptosis and tumorigenesis. , 2003, Blood.

[30]  A. D’Andrea,et al.  A novel diagnostic screen for defects in the Fanconi anemia pathway. , 2002, Blood.

[31]  A. Auerbach Fanconi anemia diagnosis and the diepoxybutane (DEB) test. , 1993, Experimental hematology.

[32]  H. Mohrenweiser,et al.  Hypomutability in Fanconi anemia cells is associated with increased deletion frequency at the HPRT locus. , 1990, Proceedings of the National Academy of Sciences of the United States of America.