Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Dev Cell ; 59(10): 1252-1268.e13, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38579720

ABSTRACT

The blueprint of the mammalian body plan is laid out during gastrulation, when a trilaminar embryo is formed. This process entails a burst of proliferation, the ingression of embryonic epiblast cells at the primitive streak, and their priming toward primitive streak fates. How these different events are coordinated remains unknown. Here, we developed and characterized a 3D culture of self-renewing mouse embryonic cells that captures the main transcriptional and architectural features of the early gastrulating mouse epiblast. Using this system in combination with microfabrication and in vivo experiments, we found that proliferation-induced crowding triggers delamination of cells that express high levels of the apical polarity protein aPKC. Upon delamination, cells become more sensitive to Wnt signaling and upregulate the expression of primitive streak markers such as Brachyury. This mechanistic coupling between ingression and differentiation ensures that the right cell types become specified at the right place during embryonic development.


Subject(s)
Cell Differentiation , Gastrulation , Germ Layers , Animals , Mice , Germ Layers/cytology , Germ Layers/metabolism , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Primitive Streak/cytology , Primitive Streak/metabolism , Fetal Proteins/metabolism , Fetal Proteins/genetics , Wnt Signaling Pathway , Cell Proliferation , Gene Expression Regulation, Developmental , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism
2.
Mol Cell ; 80(6): 996-1012.e9, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33147438

ABSTRACT

Reactive aldehydes arise as by-products of metabolism and are normally cleared by multiple families of enzymes. We find that mice lacking two aldehyde detoxifying enzymes, mitochondrial ALDH2 and cytoplasmic ADH5, have greatly shortened lifespans and develop leukemia. Hematopoiesis is disrupted profoundly, with a reduction of hematopoietic stem cells and common lymphoid progenitors causing a severely depleted acquired immune system. We show that formaldehyde is a common substrate of ALDH2 and ADH5 and establish methods to quantify elevated blood formaldehyde and formaldehyde-DNA adducts in tissues. Bone-marrow-derived progenitors actively engage DNA repair but also imprint a formaldehyde-driven mutation signature similar to aging-associated human cancer mutation signatures. Furthermore, we identify analogous genetic defects in children causing a previously uncharacterized inherited bone marrow failure and pre-leukemic syndrome. Endogenous formaldehyde clearance alone is therefore critical for hematopoiesis and in limiting mutagenesis in somatic tissues.


Subject(s)
Alcohol Dehydrogenase/genetics , Aldehyde Dehydrogenase, Mitochondrial/genetics , Formaldehyde/blood , Leukemia/genetics , Adolescent , Aldehydes/blood , Animals , Child , Child, Preschool , DNA Adducts/genetics , DNA Damage/drug effects , DNA Repair/drug effects , Female , Formaldehyde/toxicity , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Humans , Infant , Leukemia/blood , Leukemia/pathology , Male , Mice , Mutation/genetics , Substrate Specificity
3.
Nature ; 553(7687): 171-177, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29323295

ABSTRACT

Haematopoietic stem cells renew blood. Accumulation of DNA damage in these cells promotes their decline, while misrepair of this damage initiates malignancies. Here we describe the features and mutational landscape of DNA damage caused by acetaldehyde, an endogenous and alcohol-derived metabolite. This damage results in DNA double-stranded breaks that, despite stimulating recombination repair, also cause chromosome rearrangements. We combined transplantation of single haematopoietic stem cells with whole-genome sequencing to show that this damage occurs in stem cells, leading to deletions and rearrangements that are indicative of microhomology-mediated end-joining repair. Moreover, deletion of p53 completely rescues the survival of aldehyde-stressed and mutated haematopoietic stem cells, but does not change the pattern or the intensity of genome instability within individual stem cells. These findings characterize the mutation of the stem-cell genome by an alcohol-derived and endogenous source of DNA damage. Furthermore, we identify how the choice of DNA-repair pathway and a stringent p53 response limit the transmission of aldehyde-induced mutations in stem cells.


Subject(s)
Acetaldehyde/metabolism , DNA Breaks, Double-Stranded/drug effects , Ethanol/metabolism , Ethanol/pharmacology , Genomic Instability/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Mutation , Alcohol Dehydrogenase/deficiency , Alcohol Dehydrogenase/genetics , Alcohol Dehydrogenase/metabolism , Animals , Cell Survival/drug effects , DNA End-Joining Repair , Ethanol/administration & dosage , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Female , Gene Deletion , Genes, p53/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Ku Autoantigen/metabolism , Male , Mice , Mice, Inbred C57BL , Recombinational DNA Repair/drug effects , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Whole Genome Sequencing
4.
Mol Cell ; 55(6): 807-817, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25155611

ABSTRACT

Maternal metabolism provides essential nutrients to enable embryonic development. However, both mother and embryo produce reactive metabolites that can damage DNA. Here we discover how the embryo is protected from these genotoxins. Pregnant mice lacking Aldh2, a key enzyme that detoxifies reactive aldehydes, cannot support the development of embryos lacking the Fanconi anemia DNA repair pathway gene Fanca. Remarkably, transferring Aldh2(-/-)Fanca(-/-) embryos into wild-type mothers suppresses developmental defects and rescues embryonic lethality. These rescued neonates have severely depleted hematopoietic stem and progenitor cells, indicating that despite intact maternal aldehyde catabolism, fetal Aldh2 is essential for hematopoiesis. Hence, maternal and fetal aldehyde detoxification protects the developing embryo from DNA damage. Failure of this genome preservation mechanism might explain why birth defects and bone marrow failure occur in Fanconi anemia, and may have implications for fetal well-being in the many women in Southeast Asia that are genetically deficient in ALDH2.


Subject(s)
Acetaldehyde/metabolism , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Embryo, Mammalian/metabolism , Ethanol/toxicity , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia/pathology , Acetaldehyde/toxicity , Aldehyde Dehydrogenase 1 Family , Aldehyde Dehydrogenase, Mitochondrial , Animals , Animals, Newborn , DNA Damage , Disease Models, Animal , Embryo, Mammalian/embryology , Female , Genome , Hematopoietic Stem Cells/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Pregnancy , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism
5.
Mol Cell ; 54(5): 858-69, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24905007

ABSTRACT

Fanconi anaemia (FA) is a cancer predisposition syndrome characterized by cellular sensitivity to DNA interstrand crosslinkers. The molecular defect in FA is an impaired DNA repair pathway. The critical event in activating this pathway is monoubiquitination of FANCD2. In vivo, a multisubunit FA core complex catalyzes this step, but its mechanism is unclear. Here, we report purification of a native avian FA core complex and biochemical reconstitution of FANCD2 monoubiquitination. This demonstrates that the catalytic FANCL E3 ligase subunit must be embedded within the complex for maximal activity and site specificity. We genetically and biochemically define a minimal subcomplex comprising just three proteins (FANCB, FANCL, and FAAP100) that functions as the monoubiquitination module. Residual FANCD2 monoubiquitination activity is retained in cells defective for other FA core complex subunits. This work describes the in vitro reconstitution and characterization of this multisubunit monoubiquitin E3 ligase, providing key insight into the conserved FA DNA repair pathway.


Subject(s)
Avian Proteins/metabolism , Chickens/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Ubiquitination , Animals , Avian Proteins/chemistry , Avian Proteins/genetics , Cell Line , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group D2 Protein/chemistry , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group L Protein/chemistry , Fanconi Anemia Complementation Group L Protein/metabolism , Fanconi Anemia Complementation Group Proteins/chemistry , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Humans , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism
6.
EMBO J ; 32(21): 2848-60, 2013 Oct 30.
Article in English | MEDLINE | ID: mdl-24076655

ABSTRACT

RING (Really Interesting New Gene)-in-between-RING (RBR) enzymes are a distinct class of E3 ubiquitin ligases possessing a cluster of three zinc-binding domains that cooperate to catalyse ubiquitin transfer. The regulation and biological function for most members of the RBR ligases is not known, and all RBR E3s characterized to date are auto-inhibited for in vitro ubiquitylation. Here, we show that TRIAD1 and HHARI, two members of the Ariadne subfamily ligases, associate with distinct neddylated Cullin-RING ligase (CRL) complexes. In comparison to the modest E3 ligase activity displayed by isolated TRIAD1 or HHARI, binding of the cognate neddylated CRL to TRIAD1 or HHARI greatly stimulates RBR ligase activity in vitro, as determined by auto-ubiquitylation, their ability to stimulate dissociation of a thioester-linked UBCH7∼ubiquitin intermediate, and reactivity with ubiquitin-vinyl methyl ester. Moreover, genetic evidence shows that RBR ligase activity impacts both the levels and activities of neddylated CRLs in vivo. Cumulatively, our work proposes a conserved mechanism of CRL-induced Ariadne RBR ligase activation and further suggests a reciprocal role of this special class of RBRs as regulators of distinct CRLs.


Subject(s)
Carrier Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Cullin Proteins/metabolism , Cyclopentanes/pharmacology , HEK293 Cells , Humans , NEDD8 Protein , Pyrimidines/pharmacology , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination , Ubiquitins/antagonists & inhibitors , Ubiquitins/metabolism
8.
Nature ; 489(7417): 571-5, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22922648

ABSTRACT

Haematopoietic stem cells (HSCs) regenerate blood cells throughout the lifespan of an organism. With age, the functional quality of HSCs declines, partly owing to the accumulation of damaged DNA. However, the factors that damage DNA and the protective mechanisms that operate in these cells are poorly understood. We have recently shown that the Fanconi anaemia DNA-repair pathway counteracts the genotoxic effects of reactive aldehydes. Mice with combined inactivation of aldehyde catabolism (through Aldh2 knockout) and the Fanconi anaemia DNA-repair pathway (Fancd2 knockout) display developmental defects, a predisposition to leukaemia, and are susceptible to the toxic effects of ethanol-an exogenous source of acetaldehyde. Here we report that aged Aldh2(-/-) Fancd2(-/-) mutant mice that do not develop leukaemia spontaneously develop aplastic anaemia, with the concomitant accumulation of damaged DNA within the haematopoietic stem and progenitor cell (HSPC) pool. Unexpectedly, we find that only HSPCs, and not more mature blood precursors, require Aldh2 for protection against acetaldehyde toxicity. Additionally, the aldehyde-oxidizing activity of HSPCs, as measured by Aldefluor stain, is due to Aldh2 and correlates with this protection. Finally, there is more than a 600-fold reduction in the HSC pool of mice deficient in both Fanconi anaemia pathway-mediated DNA repair and acetaldehyde detoxification. Therefore, the emergence of bone marrow failure in Fanconi anaemia is probably due to aldehyde-mediated genotoxicity restricted to the HSPC pool. These findings identify a new link between endogenous reactive metabolites and DNA damage in HSCs, and define the protective mechanisms that counteract this threat.


Subject(s)
Aldehydes/toxicity , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Mutagens/toxicity , Acetaldehyde/metabolism , Acetaldehyde/toxicity , Aging , Aldehyde Dehydrogenase/deficiency , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase, Mitochondrial , Aldehydes/metabolism , Animals , Bone Marrow/pathology , DNA Damage/drug effects , DNA Damage/genetics , DNA Repair , Ethanol/toxicity , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/genetics , Female , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/metabolism , Kaplan-Meier Estimate , Leukemia/metabolism , Leukemia/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
9.
Nat Struct Mol Biol ; 18(12): 1432-4, 2011 Nov 13.
Article in English | MEDLINE | ID: mdl-22081012

ABSTRACT

Metabolism is predicted to generate formaldehyde, a toxic, simple, reactive aldehyde that can damage DNA. Here we report a synthetic lethal interaction in avian cells between ADH5, encoding the main formaldehyde-detoxifying enzyme, and the Fanconi anemia (FA) DNA-repair pathway. These results define a fundamental role for the combined action of formaldehyde catabolism and DNA cross-link repair in vertebrate cell survival.


Subject(s)
DNA Repair , Fanconi Anemia/metabolism , Formaldehyde/metabolism , Aldehyde Oxidoreductases/genetics , Animals , Cell Line , Chickens/genetics , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group C Protein/physiology , Fanconi Anemia Complementation Group L Protein/genetics , Fanconi Anemia Complementation Group L Protein/physiology , Gene Knockout Techniques , Metabolic Networks and Pathways
10.
Nature ; 475(7354): 53-8, 2011 Jul 06.
Article in English | MEDLINE | ID: mdl-21734703

ABSTRACT

Reactive aldehydes are common carcinogens. They are also by-products of several metabolic pathways and, without enzymatic catabolism, may accumulate and cause DNA damage. Ethanol, which is metabolised to acetaldehyde, is both carcinogenic and teratogenic in humans. Here we find that the Fanconi anaemia DNA repair pathway counteracts acetaldehyde-induced genotoxicity in mice. Our results show that the acetaldehyde-catabolising enzyme Aldh2 is essential for the development of Fancd2(-/-) embryos. Nevertheless, acetaldehyde-catabolism-competent mothers (Aldh2(+/-)) can support the development of double-mutant (Aldh2(-/-)Fancd2(-/-)) mice. However, these embryos are unusually sensitive to ethanol exposure in utero, and ethanol consumption by postnatal double-deficient mice rapidly precipitates bone marrow failure. Lastly, Aldh2(-/-)Fancd2(-/-) mice spontaneously develop acute leukaemia. Acetaldehyde-mediated DNA damage may critically contribute to the genesis of fetal alcohol syndrome in fetuses, as well as to abnormal development, haematopoietic failure and cancer predisposition in Fanconi anaemia patients.


Subject(s)
Aldehydes/antagonists & inhibitors , Aldehydes/toxicity , Fanconi Anemia Complementation Group D2 Protein/metabolism , Acetaldehyde/metabolism , Acetaldehyde/toxicity , Aldehyde Dehydrogenase/deficiency , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase, Mitochondrial , Aldehydes/metabolism , Alleles , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Bone Marrow/drug effects , Bone Marrow/pathology , Bone Marrow/physiopathology , Cell Line , Cell Survival/drug effects , Chickens , Clone Cells/drug effects , DNA Damage/genetics , DNA Repair/genetics , Embryo Loss/chemically induced , Embryo Loss/etiology , Embryo, Mammalian/abnormalities , Embryo, Mammalian/drug effects , Embryo, Mammalian/embryology , Ethanol/metabolism , Ethanol/toxicity , Fanconi Anemia/genetics , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/genetics , Female , Fetal Alcohol Spectrum Disorders/etiology , Gene Deletion , Genes, Essential , Hematopoiesis/drug effects , Male , Mice , Mice, Inbred C57BL , Precursor Cell Lymphoblastic Leukemia-Lymphoma/chemically induced , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , Pregnancy , Teratogens/metabolism , Teratogens/toxicity , Weaning
11.
Nat Genet ; 43(2): 147-52, 2011 02.
Article in English | MEDLINE | ID: mdl-21240276

ABSTRACT

The evolutionarily conserved SLX4 protein, a key regulator of nucleases, is critical for DNA damage response. SLX4 nuclease complexes mediate repair during replication and can also resolve Holliday junctions formed during homologous recombination. Here we describe the phenotype of the Btbd12 knockout mouse, the mouse ortholog of SLX4, which recapitulates many key features of the human genetic illness Fanconi anemia. Btbd12-deficient animals are born at sub-Mendelian ratios, have greatly reduced fertility, are developmentally compromised and are prone to blood cytopenias. Btbd12(-/-) cells prematurely senesce, spontaneously accumulate damaged chromosomes and are particularly sensitive to DNA crosslinking agents. Genetic complementation reveals a crucial requirement for Btbd12 (also known as Slx4) to interact with the structure-specific endonuclease Xpf-Ercc1 to promote crosslink repair. The Btbd12 knockout mouse therefore establishes a disease model for Fanconi anemia and genetically links a regulator of nuclease incision complexes to the Fanconi anemia DNA crosslink repair pathway.


Subject(s)
Fanconi Anemia/genetics , Recombinases/genetics , Recombinases/physiology , Animals , Cellular Senescence , Cross-Linking Reagents/pharmacology , DNA Damage , Female , Fibroblasts/metabolism , Genetic Complementation Test , Hematopoietic Stem Cells , Humans , Magnetic Resonance Imaging/methods , Male , Mice , Mice, Knockout
12.
Mol Cell ; 28(5): 798-809, 2007 Dec 14.
Article in English | MEDLINE | ID: mdl-18082605

ABSTRACT

Monoubiquitination of FANCD2 and PCNA promotes DNA repair. It causes chromatin accumulation of FANCD2 and facilitates PCNA's recruitment of translesion polymerases to stalled replication. USP1, a protease that removes monoubiquitin from FANCD2 and PCNA, was thought to reverse the DNA damage response of these substrates. We disrupted USP1 in chicken cells to dissect its role in a stable genetic system. USP1 ablation increases FANCD2 and PCNA monoubiquitination but unexpectedly results in DNA crosslinker sensitivity. This defective DNA repair is associated with constitutively chromatin-bound, monoubiquitinated FANCD2. In contrast, persistent PCNA monoubiquitination has negligible impact on DNA repair or mutagenesis. USP1 was previously shown to autocleave after DNA damage. In DT40, USP1 autocleavage is not stimulated by DNA damage, and expressing a noncleavable mutant in the USP1 knockout strain partially rescues crosslinker sensitivity. We conclude that efficient DNA crosslink repair requires FANCD2 deubiquitination, whereas FANCD2 monoubiquitination is not dependent on USP1 autocleavage.


Subject(s)
Cross-Linking Reagents/pharmacology , DNA Repair/physiology , Endopeptidases/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitination , Animals , Apoptosis , Blotting, Western , Cell Cycle , Chickens , Chromatin/metabolism , Cisplatin/pharmacology , DNA Damage/drug effects , DNA Damage/physiology , DNA Repair/drug effects , Endopeptidases/genetics , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Gene Expression Regulation , Gene Targeting , Mitomycin/pharmacology , Mutagenesis, Site-Directed , Mutation , Proliferating Cell Nuclear Antigen/genetics , Protein Processing, Post-Translational , Subcellular Fractions , Ubiquitin/metabolism , Ubiquitin-Specific Proteases
13.
Mol Cell Biol ; 27(24): 8421-30, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17938197

ABSTRACT

The Fanconi anemia (FA) nuclear core complex and the E2 ubiquitin-conjugating enzyme UBE2T are required for the S phase and DNA damage-restricted monoubiquitination of FANCD2. This constitutes a key step in the FA tumor suppressor pathway, and much attention has been focused on the regulation at this point. Here, we address the importance of the assembly of the FA core complex and the subcellular localization of UBE2T in the regulation of FANCD2 monoubiquitination. We establish three points. First, the stable assembly of the FA core complex can be dissociated of its ability to function as an E3 ubiquitin ligase. Second, the actual E3 ligase activity is not determined by the assembly of the FA core complex but rather by its DNA damage-induced localization to chromatin. Finally, UBE2T and FANCD2 access this subcellular fraction independently of the FA core complex. FANCD2 monoubiquitination is therefore not regulated by multiprotein complex assembly but by the formation of an active E2/E3 holoenzyme on chromatin.


Subject(s)
Chromatin/enzymology , Fanconi Anemia Complementation Group D2 Protein/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination , Animals , Catalytic Domain , Cell Cycle , Cell Line , Chickens , DNA Damage , Fanconi Anemia Complementation Group L Protein/metabolism , Humans , Protein Binding
14.
Nat Struct Mol Biol ; 12(9): 763-71, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16116434

ABSTRACT

The helicase-associated endonuclease for fork-structured DNA (Hef) is an archaeabacterial protein that processes blocked replication forks. Here we have isolated the vertebrate Hef ortholog and investigated its molecular function. Disruption of this gene in chicken DT40 cells results in genomic instability and sensitivity to DNA cross-links. The similarity of this phenotype to that of cells lacking the Fanconi anemia-related (FA) tumor-suppressor genes led us to investigate whether Hef functions in this pathway. Indeed, we found a genetic interaction between the FANCC and Hef genes. In addition, Hef is a component of the FA nuclear protein complex that facilitates its DNA damage-inducible chromatin localization and the monoubiquitination of the FA protein FANCD2. Notably, Hef interacts directly with DNA structures that are intermediates in DNA replication. This discovery sheds light on the origins, regulation and molecular function of the FA tumor-suppressor pathway in the maintenance of genome stability.


Subject(s)
Avian Proteins/metabolism , Cell Cycle Proteins/metabolism , Chickens , Conserved Sequence , DNA-Binding Proteins/metabolism , Fanconi Anemia/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Adenosine Triphosphatases/metabolism , Animals , Avian Proteins/chemistry , Avian Proteins/deficiency , Avian Proteins/genetics , Cell Cycle Proteins/genetics , Cell Line , Chickens/genetics , Chickens/metabolism , DNA/metabolism , DNA Damage , DNA Helicases/metabolism , DNA Repair , DNA Replication , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Endonucleases/genetics , Endonucleases/metabolism , Evolution, Molecular , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group C Protein , Fanconi Anemia Complementation Group Proteins , Genomic Instability , Humans , Nuclear Proteins/chemistry , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...