Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 171
Filter
1.
DNA Repair (Amst) ; 141: 103733, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39096698

ABSTRACT

Fanconi anemia (FA) is a hereditary disorder characterized by a deficiency in the repair of DNA interstrand crosslinks and the response to replication stress. Endogenous DNA damage, most likely caused by aldehydes, severely affects hematopoietic stem cells in FA, resulting in progressive bone marrow failure and the development of leukemia. Recent studies revealed that expression levels of SLFN11 affect the replication stress response and are a strong determinant in cell killing by DNA-damaging cancer chemotherapy. Because SLFN11 is highly expressed in the hematopoietic system, we speculated that SLFN11 may have a significant role in FA pathophysiology. Indeed, we found that DNA damage sensitivity in FA cells is significantly mitigated by the loss of SLFN11 expression. Mechanistically, we demonstrated that SLFN11 destabilizes the nascent DNA strands upon replication fork stalling. In this review, we summarize our work regarding an interplay between SLFN11 and the FA pathway, and the role of SLFN11 in the response to replication stress.


Subject(s)
DNA Damage , DNA Replication , Fanconi Anemia , Nuclear Proteins , Fanconi Anemia/metabolism , Fanconi Anemia/genetics , Humans , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Animals , DNA Repair , Fanconi Anemia Complementation Group Proteins/metabolism , Fanconi Anemia Complementation Group Proteins/genetics
2.
Commun Biol ; 6(1): 1038, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37833372

ABSTRACT

The Schlafen (SLFN)11 gene has been implicated in various biological processes such as suppression of HIV replication, replication stress response, and sensitization of cancer cells to chemotherapy. Due to the rapid diversification of the SLFN family members, it remains uncertain whether a direct ortholog of human SLFN11 exists in mice. Here we show that mSLFN8/9 and hSLFN11 were rapidly recruited to microlaser-irradiated DNA damage tracks. Furthermore, Slfn8/9 expression could complement SLFN11 loss in human SLFN11-/- cells, and as a result, reduced the growth rate to wild-type levels and partially restored sensitivity to DNA-damaging agents. In addition, both Slfn8/9 and SLFN11 expression accelerated stalled fork degradation and decreased RPA and RAD51 foci numbers after DNA damage. Based on these results, we propose that mouse Slfn8 and Slfn9 genes may share an orthologous function with human SLFN11. This notion may facilitate understanding of SLFN11's biological role through in vivo studies via mouse modeling.


Subject(s)
DNA Damage , Nuclear Proteins , Humans , Mice , Animals , Nuclear Proteins/metabolism
3.
Mol Biol Rep ; 50(10): 8385-8395, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37615925

ABSTRACT

BACKGROUND: Fanconi anemia (FA) is a devastating hereditary disorder for which we desperately need a novel therapeutic strategy. It is caused by mutations in one of at least 22 genes in the FA pathway and is characterized by developmental abnormalities, bone marrow failure, and cancer predisposition. The FA pathway is required for the efficient repair of damaged DNA, including interstrand cross-links (ICL). Recent studies indicate formaldehyde as an ultimate endogenous cause of DNA damage in FA pathophysiology. Formaldehyde can form DNA adducts as well as ICLs by inducing covalent linkages between opposite strands of double-stranded DNA. METHODS AND RESULTS: In this study, we generated a disease model of FA in zebrafish by disrupting the ube2t or fancd2 gene, which resulted in a striking phenotype of female-to-male sex reversal. Since formaldehyde is detoxified from the body by alcohol dehydrogenase 5 (ADH5), we generated fancd2-/-/adh5-/- zebrafish. We observed a body size reduction and a lower number of mature spermatozoa than wild-type or single knockout zebrafish. To evaluate if increased activity in ADH5 can affect the FA phenotype, we overexpressed human ADH5 in fancd2-/- zebrafish. The progress of spermatogenesis seemed to be partially recovered due to ADH5 overexpression. CONCLUSIONS: Our results suggest potential utility of an ADH5 enzyme activator as a therapeutic measure for the clearance of formaldehyde and treatment of FA.


Subject(s)
Fanconi Anemia , Zebrafish , Animals , Male , Humans , Female , Zebrafish/genetics , Fanconi Anemia/genetics , DNA Damage , DNA Repair , Phenotype , Formaldehyde
4.
Rinsho Ketsueki ; 64(7): 639-645, 2023.
Article in Japanese | MEDLINE | ID: mdl-37544724

ABSTRACT

Fanconi anemia (FA), a hereditary bone marrow failure syndrome, has been suggested to be caused by a defect in DNA repair that removes endogenous DNA damage due to aldehydes. In seven Japanese children with aplastic anemia who clinically resembled FA, we identified biallelic variants of the ADH5 gene, encoding formaldehyde degrading enzyme, and a heterozygous ALDH2 variant (rs671). We conclude that the combined defects in ADH5/ALDH2 caused a new disorder now termed Aldehyde Degradation Deficiency Syndrome (ADDS). We suggest that this disease is caused by defective removal of formaldehyde produced by histone demethylation during hematopoietic cell differentiation. Therapeutic targeting of formaldehyde may reduce the hematopoietic deficits of FA as well as ADDS.


Subject(s)
Fanconi Anemia , Child , Humans , Fanconi Anemia/genetics , Aldehyde Dehydrogenase, Mitochondrial/genetics , Aldehydes/metabolism , Formaldehyde , Hematopoiesis/genetics , Defense Mechanisms
5.
DNA Repair (Amst) ; 130: 103546, 2023 10.
Article in English | MEDLINE | ID: mdl-37572579

ABSTRACT

We have identified a set of Japanese children with hypoplastic anemia caused by combined defects in aldehyde degrading enzymes ADH5 and ALDH2. Their clinical characteristics overlap with a hereditary DNA repair disorder, Fanconi anemia. Our discovery of this disorder, termed Aldehyde Degradation Deficiency Syndrome (ADDS), reinforces the notion that endogenously generated aldehydes exert genotoxic effects; thus, the coupled actions of metabolism and DNA repair are required to maintain proper hematopoiesis and health.


Subject(s)
Fanconi Anemia , Child , Humans , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , DNA Repair , DNA Damage , Aldehydes/metabolism , Hematopoiesis , Aldehyde Dehydrogenase, Mitochondrial/genetics , Aldehyde Dehydrogenase, Mitochondrial/metabolism
6.
Genes Cells ; 28(9): 663-673, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37469008

ABSTRACT

The SLFN11 gene participates in cell fate decision following cancer chemotherapy and encodes the N-terminal ribonuclease (RNase) domain and the C-terminal helicase/ATPase domain. How these domains contribute to the chemotherapeutic response remains controversial. Here, we expressed SLFN11 containing mutations in two critical residues required for RNase activity in SLFN11-/- cells. We found that this mutant was still able to suppress DNA damage tolerance, destabilized the stalled replication forks, and perturbed recruitment of the fork protector RAD51. In contrast, we confirmed that the helicase domain was essential to accelerate fork degradation. The fork degradation by the RNase mutant was dependent on both DNA2 and MRE11 nuclease, but not on MRE11's novel interactor FXR1. Collectively, these results supported the view that the RNase domain function is dispensable for SLFN11 to mediate cell fate decision during replication stress response.


Subject(s)
DNA Replication , Ribonucleases , Ribonucleases/genetics , DNA Helicases/genetics , DNA Helicases/metabolism , Mutation , DNA Damage
7.
Genes Cells ; 28(9): 642-645, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37341149

ABSTRACT

Ataxia-telangiectasia (A-T) is a rare devastating hereditary condition, which affects multiple organ systems including cerebellar motor function as well as DNA repair, resulting in a higher incidence of cancer and immunodeficiency. The genetic defect in A-T lies in ATM kinase, which is activated by DNA damage and regulates a plethora of substrates including the p53 tumor suppressor. We have organized an international meeting "The 19th Ataxia-Telangiectasia Workshop 2023 (ATW2023)" with support from the Molecular Biology Society of Japan (MBSJ) and other funders. Here, we report that ATW2023 was successfully held in Kyoto from March 2nd to 5th, 2023 with more than 150 participants traveling from all over the world, despite the still smoldering COVID-19 pandemic. In this meeting report, we will briefly describe the highlights of the meeting and would like to express our gratitude to the MBSJ for the financial support.


Subject(s)
Ataxia Telangiectasia , COVID-19 , Humans , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/pathology , Ataxia Telangiectasia Mutated Proteins , Pandemics , DNA Damage , DNA Repair , Cell Cycle Proteins/metabolism
8.
Cancer Med ; 12(6): 6594-6602, 2023 03.
Article in English | MEDLINE | ID: mdl-36345163

ABSTRACT

The aldehyde degrading function of the ALDH2 enzyme is impaired by Glu504Lys polymorphisms (rs671, termed A allele), which causes alcohol flushing in east Asians, and elevates the risk of esophageal cancer among habitual drinkers. Recent studies suggested that the ALDH2 variant may lead to higher levels of DNA damage caused by endogenously generated aldehydes. This can be a threat to genome stability and/or cell viability in a synthetic manner in DNA repair-defective settings such as Fanconi anemia (FA). FA is an inherited bone marrow failure syndrome caused by defects in any one of so far identified 22 FANC genes including hereditary breast and ovarian cancer (HBOC) genes BRCA1 and BRCA2. We have previously reported that the progression of FA phenotypes is accelerated with the ALDH2 rs671 genotype. Individuals with HBOC are heterozygously mutated in either BRCA1 or BRCA2, and the cancer-initiating cells in these patients usually undergo loss of the wild-type BRCA1/2 allele, leading to homologous recombination defects. Therefore, we hypothesized that the ALDH2 genotypes may impact breast cancer development in BRCA1/2 mutant carriers. We genotyped ALDH2 in 103 HBOC patients recruited from multiple cancer centers in Japan. However, we were not able to detect any significant differences in clinical stages, histopathological classification, or age at clinical diagnosis across the ALDH2 genotypes. Unlike the effects in hematopoietic cells of FA, our current data suggest that there is no impact of the loss of ALDH2 function in cancer initiation and development in breast epithelium of HBOC patients.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial , Breast Neoplasms , Fanconi Anemia , Female , Humans , Aldehyde Dehydrogenase, Mitochondrial/genetics , BRCA1 Protein/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , East Asian People , Fanconi Anemia/genetics , Fanconi Anemia/pathology , Genetic Predisposition to Disease , Mutation , BRCA2 Protein/genetics
9.
Life Sci Alliance ; 5(12)2022 07 29.
Article in English | MEDLINE | ID: mdl-35905994

ABSTRACT

DNA damage tolerance pathways are regulated by proliferating cell nuclear antigen (PCNA) modifications at lysine 164. Translesion DNA synthesis by DNA polymerase η (Polη) is well studied, but less is known about Polη-independent mechanisms. Illudin S and its derivatives induce alkyl DNA adducts, which are repaired by transcription-coupled nucleotide excision repair (TC-NER). We demonstrate that in addition to TC-NER, PCNA modification at K164 plays an essential role in cellular resistance to these compounds by overcoming replication blockages, with no requirement for Polη. Polκ and RING finger and WD repeat domain 3 (RFWD3) contribute to tolerance, and are both dependent on PCNA modifications. Although RFWD3 is a FANC protein, we demonstrate that it plays a role in DNA damage tolerance independent of the FANC pathway. Finally, we demonstrate that RFWD3-mediated cellular survival after UV irradiation is dependent on PCNA modifications but is independent of Polη. Thus, RFWD3 contributes to PCNA modification-dependent DNA damage tolerance in addition to translesion DNA polymerases.


Subject(s)
DNA Damage , DNA-Directed DNA Polymerase , DNA Repair , DNA Replication , DNA-Directed DNA Polymerase/metabolism , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism
10.
Cell Rep ; 37(4): 109879, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34706224

ABSTRACT

SLX4/FANCP is a key Fanconi anemia (FA) protein and a DNA repair scaffold for incision around a DNA interstrand crosslink (ICL) by its partner XPF nuclease. The tandem UBZ4 ubiquitin-binding domains of SLX4 are critical for the recruitment of SLX4 to damage sites, likely by binding to K63-linked polyubiquitin chains. However, the identity of the ubiquitin E3 ligase that mediates SLX4 recruitment remains unknown. Using small interfering RNA (siRNA) screening with a GFP-tagged N-terminal half of SLX4 (termed SLX4-N), we identify the RNF168 E3 ligase as a critical factor for mitomycin C (MMC)-induced SLX4 foci formation. RNF168 and GFP-SLX4-N colocalize in MMC-induced ubiquitin foci. Accumulation of SLX4-N at psoralen-laser ICL tracks or of endogenous SLX4 at Digoxigenin-psoralen/UVA ICL is dependent on RNF168. Finally, we find that RNF168 is epistatic with SLX4 in promoting MMC tolerance. We conclude that RNF168 is a critical component of the signal transduction that recruits SLX4 to ICL damage.


Subject(s)
DNA Repair , Recombinases/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Digoxigenin/pharmacology , Ficusin/pharmacology , HCT116 Cells , Humans , MCF-7 Cells , Mitomycin/pharmacology , Recombinases/genetics , Ubiquitin/genetics , Ubiquitin-Protein Ligases/genetics
11.
Rinsho Ketsueki ; 62(6): 547-553, 2021.
Article in Japanese | MEDLINE | ID: mdl-34219079

ABSTRACT

We have recently described the identification of a novel inherited bone marrow failure syndrome. The first set of patients was diagnosed through the exome analysis of cells from Japanese patients with hypoplastic anemia, which have been deposited to the JCRB cell bank for quite some time previously. Originally, these cases were diagnosed with a novel disorder based on increased levels of sister chromatid exchanges in lymphocytes; however, causative genes were clarified only after applying the recently developed next-generation sequencing technology. Aldehyde degradation deficiency syndrome (ADDS) is caused by combined defects in two genes, ADH5 and ALDH2, which are both critical for degrading endogenously generated formaldehyde. Formaldehyde is highly reactive and toxic to biological molecules including DNA, and its endogenous generation in the absence of the degradation system results in DNA damage that overwhelms the DNA repair capacity, leading to the development of BMF with loss of hematopoietic stem cells and progression to MDS/leukemia. In this short review, we would like to summarize what is known today about ADDS for a wide readership of hematology clinicians in Japan.


Subject(s)
Fanconi Anemia , Adaptor Proteins, Signal Transducing , Alcohol Dehydrogenase , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Aldehydes , Congenital Bone Marrow Failure Syndromes , Humans , Japan
12.
Nat Struct Mol Biol ; 28(6): 487-500, 2021 06.
Article in English | MEDLINE | ID: mdl-34117478

ABSTRACT

Fanconi anemia (FA) is a devastating hereditary disease characterized by bone marrow failure (BMF) and acute myeloid leukemia (AML). As FA-deficient cells are hypersensitive to DNA interstrand crosslinks (ICLs), ICLs are widely assumed to be the lesions responsible for FA symptoms. Here, we show that FA-mutated cells are hypersensitive to persistent replication stress and that FA proteins play a role in the break-induced-replication (BIR)-like pathway for fork restart. Both the BIR-like pathway and ICL repair share almost identical molecular mechanisms of 53BP1-BRCA1-controlled signaling response, SLX4- and FAN1-mediated fork cleavage and POLD3-dependent DNA synthesis, suggesting that the FA pathway is intrinsically one of the BIR-like pathways. Replication stress not only triggers BMF in FA-deficient mice, but also specifically induces monosomy 7, which is associated with progression to AML in patients with FA, in FA-deficient cells.


Subject(s)
DNA Replication , Fanconi Anemia Complementation Group Proteins/physiology , Fanconi Anemia/genetics , Aneuploidy , Animals , Bone Marrow Failure Disorders/etiology , Cell Line, Transformed , Chickens , Chromosome Breakage , Chromosome Deletion , Chromosomes, Human, Pair 7/genetics , DNA Polymerase III/physiology , DNA Replication/genetics , Disease Progression , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group Proteins/deficiency , Fanconi Anemia Complementation Group Proteins/genetics , Female , HCT116 Cells , HEK293 Cells , Humans , Hydroxyurea/pharmacology , Leukemia, Myeloid, Acute/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Genetic , Species Specificity , Tumor Suppressor p53-Binding Protein 1/physiology , Ubiquitin-Protein Ligases/physiology
13.
Blood ; 137(15): 2021-2032, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33512438

ABSTRACT

We have recently discovered Japanese children with a novel Fanconi anemia-like inherited bone marrow failure syndrome (IBMFS). This disorder is likely caused by the loss of a catabolic system directed toward endogenous formaldehyde due to biallelic variants in ADH5 combined with a heterozygous ALDH2*2 dominant-negative allele (rs671), which is associated with alcohol-induced Asian flushing. Phytohemagglutinin-stimulated lymphocytes from these patients displayed highly increased numbers of spontaneous sister chromatid exchanges (SCEs), reflecting homologous recombination repair of formaldehyde damage. Here, we report that, in contrast, patient-derived fibroblasts showed normal levels of SCEs, suggesting that different cell types or conditions generate various amounts of formaldehyde. To obtain insights about endogenous formaldehyde production and how defects in ADH5/ALDH2 affect human hematopoiesis, we constructed disease model cell lines, including induced pluripotent stem cells (iPSCs). We found that ADH5 is the primary defense against formaldehyde, and ALDH2 provides a backup. DNA repair capacity in the ADH5/ALDH2-deficient cell lines can be overwhelmed by exogenous low-dose formaldehyde, as indicated by higher levels of DNA damage than in FANCD2-deficient cells. Although ADH5/ALDH2-deficient cell lines were healthy and showed stable growth, disease model iPSCs displayed drastically defective cell expansion when stimulated into hematopoietic differentiation in vitro, displaying increased levels of DNA damage. The expansion defect was partially reversed by treatment with a new small molecule termed C1, which is an agonist of ALDH2, thus identifying a potential therapeutic strategy for the patients. We propose that hematopoiesis or lymphocyte blastogenesis may entail formaldehyde generation that necessitates elimination by ADH5/ALDH2 enzymes.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial/genetics , Congenital Bone Marrow Failure Syndromes/genetics , Fanconi Anemia/genetics , Induced Pluripotent Stem Cells/pathology , CRISPR-Cas Systems , Cell Line , Cells, Cultured , Congenital Bone Marrow Failure Syndromes/diagnosis , Congenital Bone Marrow Failure Syndromes/pathology , DNA Damage , Fanconi Anemia/diagnosis , Fanconi Anemia/pathology , Gene Deletion , Humans , Induced Pluripotent Stem Cells/metabolism , Mutation
14.
J Radiat Res ; 62(1): 25-33, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33009557

ABSTRACT

The choice of repair pathways of DNA double-strand breaks (DSBs) is dependent upon the cell cycle phases. While homologous recombination repair (HRR) is active between the S and G2 phases, its involvement in mitotic DSB repair has not been examined in detail. In the present study, we developed a new reporter assay system to detect homology-directed repair (HDR), a major pathway used for HRR, in combination with an inducible DSB-generation system. As expected, the maximal HDR activity was observed in the late S phase, along with minimal activity in the G1 phase and at the G1/S boundary. Surprisingly, significant HDR activity was observed in M phase, and the repair efficiency was similar to that observed in late S phase. HDR was also confirmed in metaphase cells collected with continuous colcemid exposure. ChIP assays revealed the recruitment of RAD51 to the vicinity of DSBs in M phase. In addition, the ChIP assay for gamma-H2AX and phosphorylated DNA-PKcs indicated that a part of M-phase cells with DSBs could proceed into the next G1 phase. These results provide evidence showing that a portion of mitotic cell DSBs are undoubtedly repaired through action of the HDR repair pathway.


Subject(s)
DNA Breaks, Double-Stranded , Mitosis , Recombinational DNA Repair , Cell Line , Chromatin Immunoprecipitation , Humans , Kinetics , Real-Time Polymerase Chain Reaction
15.
Blood ; 137(3): 336-348, 2021 01 21.
Article in English | MEDLINE | ID: mdl-32735670

ABSTRACT

Fanconi anemia (FA) is a hereditary disorder caused by mutations in any 1 of 22 FA genes. The disease is characterized by hypersensitivity to interstrand crosslink (ICL) inducers such as mitomycin C (MMC). In addition to promoting ICL repair, FA proteins such as RAD51, BRCA2, or FANCD2 protect stalled replication forks from nucleolytic degradation during replication stress, which may have a profound impact on FA pathophysiology. Recent studies showed that expression of the putative DNA/RNA helicase SLFN11 in cancer cells correlates with cell death on chemotherapeutic treatment. However, the underlying mechanisms of SLFN11-mediated DNA damage sensitivity remain unclear. Because SLFN11 expression is high in hematopoietic stem cells, we hypothesized that SLFN11 depletion might ameliorate the phenotypes of FA cells. Here we report that SLFN11 knockdown in the FA patient-derived FANCD2-deficient PD20 cell line improved cell survival on treatment with ICL inducers. FANCD2-/-SLFN11-/- HAP1 cells also displayed phenotypic rescue, including reduced levels of MMC-induced chromosome breakage compared with FANCD2-/- cells. Importantly, we found that SLFN11 promotes extensive fork degradation in FANCD2-/- cells. The degradation process is mediated by the nucleases MRE11 or DNA2 and depends on the SLFN11 ATPase activity. This observation was accompanied by an increased RAD51 binding at stalled forks, consistent with the role of RAD51 antagonizing nuclease recruitment and subsequent fork degradation. Suppression of SLFN11 protects nascent DNA tracts even in wild-type cells. We conclude that SLFN11 destabilizes stalled replication forks, and this function may contribute to the attrition of hematopoietic stem cells in FA.


Subject(s)
DNA Replication , Fanconi Anemia/pathology , Nuclear Proteins/metabolism , Cell Cycle Checkpoints , Cell Line , Chromosome Breakage , Cross-Linking Reagents/pharmacology , DNA Helicases/metabolism , Fanconi Anemia Complementation Group D2 Protein/genetics , Gene Knockdown Techniques , Humans , MRE11 Homologue Protein/metabolism , Models, Biological , Mutation/genetics , Phenotype , RNA, Small Interfering/metabolism , Rad51 Recombinase/metabolism
16.
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
17.
PLoS One ; 15(6): e0234859, 2020.
Article in English | MEDLINE | ID: mdl-32589683

ABSTRACT

Proteins are covalently trapped on DNA to form DNA-protein cross-links (DPCs) when cells are exposed to DNA-damaging agents. Aldehyde compounds produce common types of DPCs that contain proteins in an undisrupted DNA strand. Tyrosyl-DNA phosphodiesterase 1 (TDP1) repairs topoisomerase 1 (TOPO1) that is trapped at the 3'-end of DNA. In the present study, we examined the contribution of TDP1 to the repair of formaldehyde-induced DPCs using a reverse genetic strategy with chicken DT40 cells. The results obtained showed that cells deficient in TDP1 were sensitive to formaldehyde. The removal of formaldehyde-induced DPCs was slower in tdp1-deficient cells than in wild type cells. We also found that formaldehyde did not produce trapped TOPO1, indicating that trapped TOPO1 was not a primary cytotoxic DNA lesion that was generated by formaldehyde and repaired by TDP1. The formaldehyde treatment resulted in the accumulation of chromosomal breakages that were more prominent in tdp1-deficient cells than in wild type cells. Therefore, TDP1 plays a critical role in the repair of formaldehyde-induced DPCs that are distinct from trapped TOPO1.


Subject(s)
DNA Repair , DNA Topoisomerases, Type I/metabolism , DNA/metabolism , Formaldehyde/toxicity , Phosphoric Diester Hydrolases/metabolism , Animals , Cell Line , Chickens , Chromosome Breakage/drug effects , DNA/chemistry , DNA Breaks/drug effects , DNA Breaks, Double-Stranded/drug effects , DNA Topoisomerases, Type I/chemistry , Decitabine/toxicity , Mitomycin/toxicity , Phosphoric Diester Hydrolases/genetics
18.
Oncogenesis ; 9(6): 60, 2020 Jun 15.
Article in English | MEDLINE | ID: mdl-32541651

ABSTRACT

The nucleus of mammalian cells is compartmentalized by nuclear bodies such as nuclear speckles, however, involvement of nuclear bodies, especially nuclear speckles, in DNA repair has not been actively investigated. Here, our focused screen for nuclear speckle factors involved in homologous recombination (HR), which is a faithful DNA double-strand break (DSB) repair mechanism, identified transcription-related nuclear speckle factors as potential HR regulators. Among the top hits, we provide evidence showing that USP42, which is a hitherto unidentified nuclear speckles protein, promotes HR by facilitating BRCA1 recruitment to DSB sites and DNA-end resection. We further showed that USP42 localization to nuclear speckles is required for efficient HR. Furthermore, we established that USP42 interacts with DHX9, which possesses DNA-RNA helicase activity, and is required for efficient resolution of DSB-induced R-loop. In conclusion, our data propose a model in which USP42 facilitates BRCA1 loading to DSB sites, resolution of DSB-induced R-loop and preferential DSB repair by HR, indicating the importance of nuclear speckle-mediated regulation of DSB repair.

SELECTION OF CITATIONS
SEARCH DETAIL