Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Cell Cycle ; 23(4): 339-352, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38557443

ABSTRACT

REV7 is an abundant, multifunctional protein that is a known factor in cell cycle regulation and in several key DNA repair pathways including Trans-Lesion Synthesis (TLS), the Fanconi Anemia (FA) pathway, and DNA Double-Strand Break (DSB) repair pathway choice. Thus far, no direct role has been studied for REV7 in the DNA damage response (DDR) signaling pathway. Here we describe a novel function for REV7 in DSB-induced p53 signaling. We show that REV7 binds directly to p53 to block ATM-dependent p53 Ser15 phosphorylation. We also report that REV7 is involved in the destabilization of p53. These findings affirm REV7's participation in fundamental cell cycle and DNA repair pathways. Furthermore, they highlight REV7 as a critical factor for the integration of multiple processes that determine viability and genome stability.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , DNA Damage , Signal Transduction , Tumor Suppressor Protein p53 , Ataxia Telangiectasia Mutated Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Humans , Phosphorylation , DNA Breaks, Double-Stranded , Protein Binding , DNA Repair , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Cell Line, Tumor
2.
CJC Pediatr Congenit Heart Dis ; 2(1): 51-54, 2023 Feb.
Article in English | MEDLINE | ID: mdl-37970106

ABSTRACT

Atrial fibrillation (AF) can occur predominantly associated with right atrial (RA) lesions in congenital heart disease, particularly when the RA cavity is dilated. RA electrical potentials occasionally appear organized during AF. We clearly mapped such areas circumscribed by an intra-atrial re-entrant circuit during an isoproterenol infusion, in a patient with a repaired tetralogy of Fallot, using an ultrahigh-density mapping system and its beat acceptance criteria function. Ablation of areas inside the re-entrant circuit successfully eliminated the AF. Our experience indicated that a macro-re-entrant tachycardia was a driver as well as a trigger of AF of this right-sided origin.


Chez les patients atteints d'une cardiopathie congénitale, la fibrillation auriculaire (FA) peut souvent survenir en association avec des lésions auriculaires droites (AD), en particulier lorsque la cavité AD est dilatée. Lors d'une FA, il peut arriver que les potentiels électriques AD semblent normaux. Chez un patient ayant une tétralogie de Fallot réparée, nous avons clairement cartographié des zones délimitées par un circuit de réentrée intra-auriculaire lors d'une perfusion d'isoprotérénol, et ce, à l'aide d'un système de cartographie à très haute densité et de ses critères d'acceptation liés aux battements cardiaques. L'ablation des régions se trouvant dans le circuit de réentrée a permis d'éliminer la FA avec succès. Notre expérience a démontré qu'une tachycardie macroréentrante avait été un facteur déterminant et même un déclencheur de la FA, laquelle est apparue à droite.

3.
EMBO J ; 40(21): e104543, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34533226

ABSTRACT

The DNA polymerase zeta (Polζ) plays a critical role in bypassing DNA damage. REV3L, the catalytic subunit of Polζ, is also essential in mouse embryonic development and cell proliferation for reasons that remain incompletely understood. In this study, we reveal that REV3L protein interacts with heterochromatin components including repressive histone marks and localizes in pericentromeric regions through direct interaction with HP1 dimer. We demonstrate that Polζ/REV3L ensures progression of replication forks through difficult-to-replicate pericentromeric heterochromatin, thereby preventing spontaneous chromosome break formation. We also find that Rev3l-deficient cells are compromised in the repair of heterochromatin-associated double-stranded breaks, eliciting deletions in late-replicating regions. Lack of REV3L leads to further consequences that may be ascribed to heterochromatin replication and repair-associated functions of Polζ, with a disruption of the temporal replication program at specific loci. This is correlated with changes in epigenetic landscape and transcriptional control of developmentally regulated genes. These results reveal a new function of Polζ in preventing chromosome instability during replication of heterochromatic regions.


Subject(s)
DNA Replication , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , DNA/genetics , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Heterochromatin/metabolism , Animals , Cell Line , Cell Line, Transformed , Cell Proliferation , Chromobox Protein Homolog 5/genetics , Chromobox Protein Homolog 5/metabolism , Chromosomal Instability , DNA/metabolism , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Embryo, Mammalian , Embryonic Stem Cells/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , HeLa Cells , Heterochromatin/chemistry , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , NIH 3T3 Cells , Signal Transduction
4.
Cell Rep ; 34(8): 108775, 2021 02 23.
Article in English | MEDLINE | ID: mdl-33626348

ABSTRACT

In mammalian cells, specialized DNA polymerase ζ (pol ζ) contributes to genomic stability during normal DNA replication. Disruption of the catalytic subunit Rev3l is toxic and results in constitutive chromosome damage, including micronuclei. As manifestations of this genomic stress are unknown, we examined the transcriptome of pol ζ-defective cells by RNA sequencing (RNA-seq). Expression of 1,117 transcripts is altered by ≥4-fold in Rev3l-disrupted cells, with a pattern consistent with an induction of an innate immune response. Increased expression of interferon-stimulated genes at the mRNA and protein levels in pol ζ-defective cells is driven by the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-signaling partner stimulator of interferon genes (STING) pathway. Expression of key interferon-stimulated chemokines is elevated in basal epithelial mouse skin cells with a disruption of Rev3l. These results indicate that the disruption of pol ζ may simultaneously increase sensitivity to genotoxins and potentially engage parts of the innate immune response, which could add an additional benefit to targeting pol ζ in cancer therapies.


Subject(s)
DNA Damage , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Fibroblasts/enzymology , Genomic Instability , Immunity, Innate , Micronuclei, Chromosome-Defective , Animals , Cell Line , Chemokines/genetics , Chemokines/metabolism , Cisplatin/pharmacology , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , Fibroblasts/drug effects , Fibroblasts/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Knockout , Micronuclei, Chromosome-Defective/chemically induced , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Signal Transduction , Transcriptome
5.
Microorganisms ; 9(1)2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33374596

ABSTRACT

Plasmodium vivax malaria is a neglected tropical disease, despite being more geographically widespread than any other form of malaria. The documentation of P. vivax infections in different parts of Africa where Duffy-negative individuals are predominant suggested that there are alternative pathways for P. vivax to invade human erythrocytes. Duffy-negative individuals may be just as fit as Duffy-positive individuals and are no longer resistant to P.vivax malaria. In this review, we describe the complexity of P. vivax malaria, characterize pathogenesis and candidate invasion genes of P. vivax, and host immune responses to P. vivax infections. We provide a comprehensive review on parasite ligands in several Plasmodium species that further justify candidate genes in P. vivax. We also summarize previous genomic and transcriptomic studies related to the identification of ligand and receptor proteins in P. vivax erythrocyte invasion. Finally, we identify topics that remain unclear and propose future studies that will greatly contribute to our knowledge of P. vivax.

6.
Environ Mol Mutagen ; 61(7): 709-715, 2020 08.
Article in English | MEDLINE | ID: mdl-32306447

ABSTRACT

FAM35A, alternatively known as SHLD2 and RINN2, was recently characterized as a DNA repair gene, evolutionarily conserved in higher vertebrates. FAM35A is a 53BP1-pathway factor and a component of the Shieldin/RINN complex. Among 53BP1-pathway factors, FAM35A has unique domains: an N-terminal disordered domain and three C-terminal OB-fold domains. These C-terminal domains have homology with the OB-fold domains of the single-stranded DNA binding protein, RPA1. With other 53BP1-pathway factors, FAM35A inhibits DNA end resection. FAM35A defective cell lines are sensitive to DNA double-strand break inducing agents. Concurrent FAM35A and BRCA1 defects in mammalian cell lines cause resistance to PARP inhibitors and camptothecin. The clinical relevance of this interaction is still unknown, but cancer genomics databases indicate that FAM35A is deleted in 6-13% of prostate cancers and in at least one triple negative breast cancer patient-derived BRCA1 defective cell line. From meta-analysis, FAM35A overexpression in patients with triple negative and basal-like breast cancers is associated with poor survival compared to patients with low expression. From this evidence, clarification of FAM35A's function and the related mechanism of chemoresistance is likely to have clinical implications.


Subject(s)
DNA Repair/genetics , DNA-Binding Proteins/genetics , Genomic Instability/genetics , Neoplasms/genetics , Animals , DNA Breaks, Double-Stranded , Humans
7.
J Phys Chem Lett ; 11(9): 3345-3349, 2020 May 07.
Article in English | MEDLINE | ID: mdl-32294382

ABSTRACT

Serum albumin-gold complexes exhibit UV-excitable red luminescence (λem = 640 nm) with unusual Stokes shifts compared with the innate UV/blue fluorescence arising from the aromatic residues. In order to understand the mechanism of this luminescence, we employed limited proteolysis and molecular cloning techniques and assessed the domain containing the red luminophore in bovine serum albumin (BSA) and human serum albumin (HSA). We identified that the luminophore is localized in a domain of serum albumin, residing within the N-terminus half.


Subject(s)
Gold/chemistry , Serum Albumin, Bovine/chemistry , Serum Albumin, Human/chemistry , Luminescence , Protein Domains
8.
FEBS J ; 286(1): 139-150, 2019 01.
Article in English | MEDLINE | ID: mdl-30431240

ABSTRACT

R-loops, which consist of DNA : RNA hybrids and displaced single-strand DNA, are a major threat to genome stability. We have previously reported that a key Fanconi anemia protein, FANCD2, accumulates on large fragile genes during mild replication stress in a manner depending on R-loops. In this study, we found that FANCD2 suppresses R-loop levels. Furthermore, we identified FANCD2 interactions with RNA processing factors, including hnRNP U and DDX47. Our data suggest that FANCD2, which accumulates with R-loops in chromatin, recruits these factors and thereby promotes efficient processing of long RNA transcripts. This may lead to a reduction in transcription-replication collisions, as detected by PLA between PCNA and RNA Polymerase II, and hence, lowered R-loop levels. We propose that this mechanism might contribute to maintenance of genome stability during mild replication stress.


Subject(s)
DEAD-box RNA Helicases/metabolism , DNA Replication , Fanconi Anemia Complementation Group D2 Protein/metabolism , Genomic Instability , Heterogeneous-Nuclear Ribonucleoprotein U/metabolism , RNA Polymerase II/metabolism , RNA, Neoplasm/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , DEAD-box RNA Helicases/genetics , DNA Repair , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Fanconi Anemia Complementation Group D2 Protein/genetics , Heterogeneous-Nuclear Ribonucleoprotein U/genetics , Humans , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , RNA Polymerase II/genetics , RNA Processing, Post-Transcriptional , RNA, Neoplasm/chemistry , RNA, Neoplasm/genetics , Tumor Cells, Cultured
9.
EMBO J ; 37(12)2018 06 15.
Article in English | MEDLINE | ID: mdl-29789392

ABSTRACT

To exploit vulnerabilities of tumors, it is urgent to identify associated defects in genome maintenance. One unsolved problem is the mechanism of regulation of DNA double-strand break repair by REV7 in complex with 53BP1 and RIF1, and its influence on repair pathway choice between homologous recombination and non-homologous end-joining. We searched for REV7-associated factors in human cells and found FAM35A, a previously unstudied protein with an unstructured N-terminal region and a C-terminal region harboring three OB-fold domains similar to single-stranded DNA-binding protein RPA, as novel interactor of REV7/RIF1/53BP1. FAM35A re-localized in damaged cell nuclei, and its knockdown caused sensitivity to DNA-damaging agents. In a BRCA1-mutant cell line, however, depletion of FAM35A increased resistance to camptothecin, suggesting that FAM35A participates in processing of DNA ends to allow more efficient DNA repair. We found FAM35A absent in one widely used BRCA1-mutant cancer cell line (HCC1937) with anomalous resistance to PARP inhibitors. A survey of FAM35A alterations revealed that the gene is altered at the highest frequency in prostate cancers (up to 13%) and significantly less expressed in metastatic cases, revealing promise for FAM35A as a therapeutically relevant cancer marker.


Subject(s)
BRCA1 Protein/deficiency , Biomarkers, Tumor/metabolism , DNA Damage , DNA Repair , DNA, Neoplasm/metabolism , Mad2 Proteins/metabolism , Neoplasms/metabolism , Proteins/metabolism , Biomarkers, Tumor/genetics , Cell Cycle Proteins , Cell Line, Tumor , DNA, Neoplasm/genetics , DNA-Binding Proteins , HEK293 Cells , Humans , Mad2 Proteins/genetics , Mutation , Neoplasms/genetics , Neoplasms/pathology , Proteins/genetics , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism
10.
Mutat Res ; 803-805: 89-95, 2017 10.
Article in English | MEDLINE | ID: mdl-28552166

ABSTRACT

Fanconi anemia (FA) is a devastating hereditary condition that impacts genome integrity, leading to clinical features such as skeletal and visceral organ malformations, attrition of bone marrow stem cells, and carcinogenesis. At least 21 proteins, when absent or defective, have been implicated in this disorder, and they together constitute the FA pathway, which functions in detection and repair of, and tolerance to, endogenous DNA damage. The damage primarily handled by the FA pathway has been assumed to be related to DNA interstrand crosslinks (ICLs). The FA pathway is activated upon ICL damage, and a hallmark of this activation is the mono-ubiquitination events of the key FANCD2-FANCI protein complex. Recent data have revealed unexpectedly complex details in the regulation of FA pathway activation by ICLs. In this short review, we summarize the knowledge accumulated over the years regarding how the FA pathway is activated via protein modifications.


Subject(s)
DNA Damage , Fanconi Anemia/genetics , Protein Processing, Post-Translational , Ubiquitination , Animals , Cell Line, Tumor , Chickens , DNA Repair , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Fibroblasts/cytology , HCT116 Cells , Humans , Phosphorylation , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism
11.
Proc Natl Acad Sci U S A ; 113(48): 13774-13779, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27849610

ABSTRACT

Routine dietary consumption of foods that contain aflatoxins is the second leading cause of environmental carcinogenesis worldwide. Aflatoxin-driven mutagenesis is initiated through metabolic activation of aflatoxin B1 (AFB1) to its epoxide form that reacts with N7 guanine in DNA. The resulting AFB1-N7-dG adduct undergoes either spontaneous depurination or imidazole-ring opening yielding formamidopyrimidine AFB1 (AFB1-Fapy-dG). Because this latter adduct is known to persist in human tissues and contributes to the high frequency G-to-T mutation signature associated with many hepatocellular carcinomas, we sought to establish the identity of the polymerase(s) involved in processing this lesion. Although our previous biochemical analyses demonstrated the ability of polymerase ζ (pol ζ) to incorporate an A opposite AFB1-Fapy-dG and extend from this mismatch, biological evidence supporting a unique role for this polymerase in cellular tolerance following aflatoxin exposure has not been established. Following challenge with AFB1, survival of mouse cells deficient in pol ζ (Rev3L-/-) was significantly reduced relative to Rev3L+/- cells or Rev3L-/- cells complemented through expression of the wild-type human REV3L. Furthermore, cell-cycle progression of Rev3L-/- mouse embryo fibroblasts was arrested in late S/G2 following AFB1 exposure. These Rev3L-/- cells showed an increase in replication-dependent formation of γ-H2AX foci, micronuclei, and chromosomal aberrations (chromatid breaks and radials) relative to Rev3L+/- cells. These data suggest that pol ζ is essential for processing AFB1-induced DNA adducts and that, in its absence, cells do not have an efficient backup polymerase or a repair/tolerance mechanism facilitating survival.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , Liver Neoplasms/genetics , Aflatoxin B1/analogs & derivatives , Aflatoxin B1/genetics , Aflatoxin B1/toxicity , Aflatoxins/toxicity , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/pathology , Cell Survival/drug effects , Chromosome Aberrations/drug effects , Cytidine/analogs & derivatives , Cytidine/genetics , Cytidine/toxicity , DNA Adducts/drug effects , DNA Adducts/genetics , DNA Damage/drug effects , DNA Repair/genetics , DNA-Directed DNA Polymerase/chemistry , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms/pathology , Mice , Mutagenesis/drug effects , Mutagenesis/genetics , Mutation
12.
Genes Dev ; 30(10): 1187-97, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27198228

ABSTRACT

Histone H3 methylation on Lys4 (H3K4me) is associated with active gene transcription in all eukaryotes. In Saccharomyces cerevisiae, Set1 is the sole lysine methyltransferase required for mono-, di-, and trimethylation of this site. Although H3K4me3 is linked to gene expression, whether H3K4 methylation regulates other cellular processes, such as mitosis, is less clear. Here we show that both Set1 and H3K4 mutants display a benomyl resistance phenotype that requires components of the spindle assembly checkpoint (SAC), including Bub3 and Mad2. These proteins inhibit Cdc20, an activator of the anaphase-promoting complex/cyclosome (APC/C). Mutations in Cdc20 that block Mad2 interactions suppress the benomyl resistance of both set1 and H3K4 mutant cells. Furthermore, the HORMA domain in Mad2 directly binds H3, identifying a new histone H3 "reader" motif. Mad2 undergoes a conformational change important for execution of the SAC. We found that the closed (active) conformation of both yeast and human Mad2 is capable of binding methylated H3K4, but, in contrast, the open (inactive) Mad2 conformation limits interaction with methylated H3. Collectively, our data indicate that interactions between Mad2 and H3K4 regulate resolution of the SAC by limiting closed Mad2 availability for Cdc20 inhibition.


Subject(s)
Histones/metabolism , M Phase Cell Cycle Checkpoints/genetics , Mad2 Proteins/metabolism , Benomyl/pharmacology , Cdc20 Proteins/genetics , Cdc20 Proteins/metabolism , Drug Resistance/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Humans , M Phase Cell Cycle Checkpoints/drug effects , Methylation , Mutation , Protein Binding/genetics , Protein Conformation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Spindle Apparatus/genetics , Spindle Apparatus/pathology , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , Tubulin Modulators/pharmacology
13.
PLoS Genet ; 12(1): e1005759, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26727495

ABSTRACT

DNA polymerase ζ (pol ζ) is exceptionally important for maintaining genome stability. Inactivation of the Rev3l gene encoding the polymerase catalytic subunit causes a high frequency of chromosomal breaks, followed by lethality in mouse embryos and in primary cells. Yet it is not known whether the DNA polymerase activity of pol ζ is specifically essential, as the large REV3L protein also serves as a multiprotein scaffold for translesion DNA synthesis via multiple conserved structural domains. We report that Rev3l cDNA rescues the genomic instability and DNA damage sensitivity of Rev3l-null immortalized mouse fibroblast cell lines. A cDNA harboring mutations of conserved catalytic aspartate residues in the polymerase domain of REV3L could not rescue these phenotypes. To investigate the role of REV3L DNA polymerase activity in vivo, a Rev3l knock-in mouse was constructed with this polymerase-inactivating alteration. No homozygous mutant mice were produced, with lethality occurring during embryogenesis. Primary fibroblasts from mutant embryos showed growth defects, elevated DNA double-strand breaks and cisplatin sensitivity similar to Rev3l-null fibroblasts. We tested whether the severe Rev3l-/- phenotypes could be rescued by deletion of DNA polymerase η, as has been reported with chicken DT40 cells. However, Rev3l-/- Polh-/- mice were inviable, and derived primary fibroblasts were as sensitive to DNA damage as Rev3l-/- Polh+/+ fibroblasts. Therefore, the functions of REV3L in maintaining cell viability, embryonic viability and genomic stability are directly dependent on its polymerase activity, and cannot be ameliorated by an additional deletion of pol η. These results validate and encourage the approach of targeting the DNA polymerase activity of pol ζ to sensitize tumors to DNA damaging agents.


Subject(s)
Cell Survival/genetics , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , Embryonic Development/genetics , Animals , DNA Breaks, Double-Stranded , DNA Damage/genetics , DNA Replication/genetics , Embryo, Mammalian , Gene Knock-In Techniques , Genomic Instability , Mice , Mutation
14.
J Biol Chem ; 290(40): 24278-93, 2015 Oct 02.
Article in English | MEDLINE | ID: mdl-26269593

ABSTRACT

DNA polymerase ν (POLN) is one of 16 DNA polymerases encoded in vertebrate genomes. It is important to determine its gene expression patterns, biological roles, and biochemical activities. By quantitative analysis of mRNA expression, we found that POLN from the zebrafish Danio rerio is expressed predominantly in testis. POLN is not detectably expressed in zebrafish embryos or in mouse embryonic stem cells. Consistent with this, injection of POLN-specific morpholino antisense oligonucleotides did not interfere with zebrafish embryonic development. Analysis of transcripts revealed that vertebrate POLN has an unusual gene expression arrangement, sharing a first exon with HAUS3, the gene encoding augmin-like complex subunit 3. HAUS3 is broadly expressed in embryonic and adult tissues, in contrast to POLN. Differential expression of POLN and HAUS3 appears to arise by alternate splicing of transcripts in mammalian cells and zebrafish. When POLN was ectopically overexpressed in human cells, it specifically coimmunoprecipitated with the homologous recombination factors BRCA1 and FANCJ, but not with previously suggested interaction partners (HELQ and members of the Fanconi anemia core complex). Purified zebrafish POLN protein is capable of thymine glycol bypass and strand displacement, with activity dependent on a basic amino acid residue known to stabilize the primer-template. These properties are conserved with the human enzyme. Although the physiological function of pol ν remains to be clarified, this study uncovers distinctive aspects of its expression control and evolutionarily conserved properties of this DNA polymerase.


Subject(s)
DNA-Directed DNA Polymerase/metabolism , Gene Expression Regulation , Testis/metabolism , Zebrafish Proteins/metabolism , Alternative Splicing , Animals , BRCA1 Protein/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , DNA/chemistry , DNA Damage , Exons , Fanconi Anemia Complementation Group Proteins/metabolism , Gene Order , Genes, Overlapping , HEK293 Cells , HeLa Cells , Humans , Male , Mice , Recombination, Genetic , Transgenes , Zebrafish
15.
Nucleic Acids Res ; 43(2): 1000-11, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25567983

ABSTRACT

DNA polymerase zeta (pol ζ) is exceptionally important for controlling mutagenesis and genetic instability. REV3L comprises the catalytic subunit, while REV7 (MAD2L2) is considered an accessory subunit. However, it has not been established that the role of REV7 in DNA damage tolerance is necessarily connected with mammalian pol ζ, and there is accumulating evidence that REV7 and REV3L have independent functions. Analysis of pol ζ has been hampered by difficulties in expression of REV3L in mammalian cells, and lack of a functional complementation system. Here, we report that REV7 interacts with full-length REV3L in vivo and we identify a new conserved REV7 interaction site in human REV3L (residues 1993-2003), distinct from the known binding site (residues 1877-1887). Mutation of both REV7-binding sites eliminates the REV3L-REV7 interaction. In vivo complementation shows that both REV7-binding sites in REV3L are necessary for preventing spontaneous chromosome breaks and conferring resistance to UV radiation and cisplatin. This demonstrates a damage-specific function of REV7 in pol ζ, in contrast to the distinct roles of REV3L and REV7 in primary cell viability and embryogenesis.


Subject(s)
DNA Damage , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Mad2 Proteins/metabolism , Binding Sites , Cell Line , DNA-Binding Proteins/chemistry , DNA-Directed DNA Polymerase/chemistry , HeLa Cells , Humans
16.
PLoS Genet ; 10(10): e1004654, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25275444

ABSTRACT

Although a defect in the DNA polymerase POLQ leads to ionizing radiation sensitivity in mammalian cells, the relevant enzymatic pathway has not been identified. Here we define the specific mechanism by which POLQ restricts harmful DNA instability. Our experiments show that Polq-null murine cells are selectively hypersensitive to DNA strand breaking agents, and that damage resistance requires the DNA polymerase activity of POLQ. Using a DNA break end joining assay in cells, we monitored repair of DNA ends with long 3' single-stranded overhangs. End joining events retaining much of the overhang were dependent on POLQ, and independent of Ku70. To analyze the repair function in more detail, we examined immunoglobulin class switch joining between DNA segments in antibody genes. POLQ participates in end joining of a DNA break during immunoglobulin class-switching, producing insertions of base pairs at the joins with homology to IgH switch-region sequences. Biochemical experiments with purified human POLQ protein revealed the mechanism generating the insertions during DNA end joining, relying on the unique ability of POLQ to extend DNA from minimally paired primers. DNA breaks at the IgH locus can sometimes join with breaks in Myc, creating a chromosome translocation. We found a marked increase in Myc/IgH translocations in Polq-defective mice, showing that POLQ suppresses genomic instability and genome rearrangements originating at DNA double-strand breaks. This work clearly defines a role and mechanism for mammalian POLQ in an alternative end joining pathway that suppresses the formation of chromosomal translocations. Our findings depart from the prevailing view that alternative end joining processes are generically translocation-prone.


Subject(s)
Chromosomal Instability , DNA-Directed DNA Polymerase/metabolism , Animals , B-Lymphocytes/physiology , Bleomycin/pharmacology , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Bone Marrow Cells/radiation effects , Cells, Cultured , DNA Breaks, Double-Stranded , DNA End-Joining Repair/genetics , DNA-Directed DNA Polymerase/genetics , Female , HEK293 Cells , Humans , Immunoglobulin Class Switching , Metabolic Networks and Pathways , Mice, Inbred C57BL , Mice, Mutant Strains , DNA Polymerase theta
17.
Cell Rep ; 7(4): 1039-47, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24794430

ABSTRACT

The Fanconi anemia (FA) pathway is critically involved in the maintenance of hematopoietic stem cells and the suppression of carcinogenesis. A key FA protein, FANCD2, is monoubiquitinated and accumulates in chromatin in response to DNA interstrand crosslinks (ICLs), where it coordinates DNA repair through mechanisms that are still poorly understood. Here, we report that CtIP protein directly interacts with FANCD2. A region spanning amino acids 166 to 273 of CtIP and monoubiquitination of FANCD2 are both essential for the FANCD2-CtIP interaction and mitomycin C (MMC)-induced CtIP foci. Remarkably, both FANCD2 and CtIP are critical for MMC-induced RPA2 hyperphosphorylation, an event that accompanies end resection of double-strand breaks. Collectively, our results reveal a role of monoubiquitinated FANCD2 in end resection that depends on its binding to CtIP during ICL repair.


Subject(s)
Carrier Proteins/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia/genetics , Nuclear Proteins/genetics , Carrier Proteins/metabolism , Cross-Linking Reagents , DNA Breaks, Double-Stranded , DNA Damage , DNA Repair , Endodeoxyribonucleases , Fanconi Anemia Complementation Group D2 Protein/metabolism , Humans , Nuclear Proteins/metabolism , Transfection
18.
Nat Commun ; 4: 2338, 2013.
Article in English | MEDLINE | ID: mdl-24005565

ABSTRACT

Mammalian HELQ is a 3'-5' DNA helicase with strand displacement activity. Here we show that HELQ participates in a pathway of resistance to DNA interstrand crosslinks (ICLs). Genetic disruption of HELQ in human cells enhances cellular sensitivity and chromosome radial formation by the ICL-inducing agent mitomycin C (MMC). A significant fraction of MMC sensitivity is independent of the Fanconi anaemia pathway. Sister chromatid exchange frequency and sensitivity to UV radiation or topoisomerase inhibitors is unaltered. Proteomic analysis reveals that HELQ is associated with the RAD51 paralogs RAD51B/C/D and XRCC2, and with the DNA damage-responsive kinase ATR. After treatment with MMC, reduced phosphorylation of the ATR substrate CHK1 occurs in HELQ-knockout cells, and accumulation of G2/M cells is reduced. The results indicate that HELQ operates in an arm of DNA repair and signalling in response to ICL. Further, the association with RAD51 paralogs suggests HELQ as a candidate ovarian cancer gene.


Subject(s)
Cross-Linking Reagents/pharmacology , DNA Helicases/metabolism , DNA/metabolism , Rad51 Recombinase/metabolism , Sequence Homology, Amino Acid , Ataxia Telangiectasia Mutated Proteins/metabolism , Base Sequence , Cell Line , Checkpoint Kinase 1 , DNA Copy Number Variations/genetics , DNA Damage , Enzyme Activation/drug effects , Fanconi Anemia/genetics , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group D2 Protein/genetics , Female , Gene Knockout Techniques , HEK293 Cells , HeLa Cells , Humans , Mitomycin/pharmacology , Molecular Sequence Data , Mutant Proteins/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Protein Binding/drug effects , Protein Kinases/metabolism , Sister Chromatid Exchange/drug effects
19.
Nucleic Acids Res ; 41(14): 6930-41, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23723247

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , DNA Repair , DNA-Binding Proteins/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/analysis , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/physiology , Cell Line , Chromatin/chemistry , DNA Replication , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/analysis , DNA-Binding Proteins/physiology , HeLa Cells , Humans , Replication Protein A/metabolism
20.
Cancer Res ; 72(5): 1149-56, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22258451

ABSTRACT

ATR kinase activates the S-phase checkpoint when replication forks stall at sites of DNA damage. This event also causes phosphorylation of the Fanconi anemia (FA) protein FANCI, triggering its monoubiquitination of the key DNA repair factor FANCD2 by the FA core E3 ligase complex, thereby promoting this central pathway of DNA repair which permits replication to be restarted. However, the interplay between ATR and the FA pathway has been unclear. In this study, we present evidence that their action is directly linked, gaining insights into this relationship in a DT40 mutant cell line that is conditionally deficient in the critical ATR-binding partner protein ATRIP. Using this system, we showed that ATRIP was crucial for DNA damage-induced FANCD2 monoubiquitination and FANCI phosphorylation. ATR kinase phosphorylated recombinant FANCI protein in vitro, which was facilitated by the presence of FANCD2. Mechanistic investigations revealed that the RPA region but not the TopBP1 region of ATRIP was required for FANCD2 monoubiquitination, whereas Chk1 phosphorylation relied upon both domains. Together, our findings identify ATR as the kinase responsible for activating the FA pathway of DNA repair.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/physiology , DNA Repair , DNA-Binding Proteins/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Protein Serine-Threonine Kinases/physiology , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Line , Chickens , Fanconi Anemia Complementation Group D2 Protein/metabolism , Humans , Mutation , Recombinant Proteins , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...