Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.335
Filter
1.
Biochem Biophys Res Commun ; 712-713: 149893, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38657529

ABSTRACT

RecJ exonucleases are members of the DHH phosphodiesterase family ancestors of eukaryotic Cdc45, the key component of the CMG (Cdc45-MCM-GINS) complex at the replication fork. They are involved in DNA replication and repair, RNA maturation and Okazaki fragment degradation. Bacterial RecJs resect 5'-end ssDNA. Conversely, archaeal RecJs are more versatile being able to hydrolyse in both directions and acting on ssDNA as well as on RNA. In Methanocaldococcus jannaschii two RecJs were previously characterized: RecJ1 is a 5'→3' DNA exonuclease, MjaRecJ2 works only on 3'-end DNA/RNA with a preference for RNA. Here, I present the crystal structure of MjaRecJ2, solved at a resolution of 2.8 Å, compare it with the other RecJ structures, in particular the 5'→3' TkoGAN and the bidirectional PfuRecJ, and discuss its characteristics in light of the more recent knowledge on RecJs. This work adds new structural data that might improve the knowledge of these class of proteins.


Subject(s)
Methanocaldococcus , Models, Molecular , Methanocaldococcus/enzymology , Crystallography, X-Ray , Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , Archaeal Proteins/genetics , Exonucleases/metabolism , Exonucleases/chemistry , Protein Conformation , Amino Acid Sequence , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/metabolism , Exodeoxyribonucleases/genetics
2.
Viruses ; 16(4)2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38675842

ABSTRACT

TREX1 acts in the initial prevention of an autoimmune response, but it may contribute to the permissiveness of retrovirus infections. This study investigated the association between the levels of TREX1 gene expression with the polymorphisms TREX1 rs3135941 (T/C) and TREX1 rs3135945 (G/A), and the presence of antinuclear antibodies (ANA) in antiretroviral therapy (ART)-naïve individuals and after 1 year of treatment. Blood samples from 119 individuals with HIV-1 were subjected to genotyping of polymorphisms and quantification of TREX1 gene expression and HIV-1 viral load by qPCR. The concentration of IFN-α and the number of CD4+/CD8+ T lymphocytes were determined by ELISA and flow cytometry, respectively; ANA was investigated by immunofluorescence. A control group of 167 seronegative individuals was used for the comparison of genotypic frequencies. The frequency of the polymorphisms were not associated with HIV infection or with variations in the expression of TREX1 and IFN-α (p > 0.05). ART-naïve individuals exhibited higher TREX1 expression and lower IFN-α expression. After 1 year of ART, TREX1 levels were reduced, while IFN-α and CD4+ T lymphocytes were elevated (p < 0.05). Some individuals on ART presented ANA. These results suggest that ART-mediated restoration of immune competence is associated with a reduction in TREX1 expression, which may induce the development of ANA, regardless of the polymorphism investigated.


Subject(s)
Exodeoxyribonucleases , HIV Infections , HIV-1 , Immune Reconstitution , Phosphoproteins , Adult , Female , Humans , Male , Middle Aged , Antibodies, Antinuclear/blood , CD4-Positive T-Lymphocytes/immunology , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Genotype , HIV Infections/immunology , HIV Infections/drug therapy , HIV Infections/genetics , HIV Infections/virology , HIV-1/immunology , Immune Reconstitution/genetics , Immune Reconstitution/immunology , Interferon-alpha/blood , Interferon-alpha/metabolism , Phosphoproteins/genetics , Polymorphism, Single Nucleotide , Viral Load , Anti-Retroviral Agents/adverse effects , Anti-Retroviral Agents/therapeutic use
3.
Talanta ; 274: 125934, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38574533

ABSTRACT

Nowadays, novel and efficient signal amplification strategy in electrochemiluminescence (ECL) platform is urgently needed to enhance the sensitivity of biosensor. In this work, the dual ECL signal enhancement strategy was constructed by the interactions of Pd nanoparticles attached covalent organic frameworks (Pd NPs@COFs) with tris (bipyridine) ruthenium (RuP) and Exonuclease III (Exo.III) cycle reaction. Within this strategy, the COFs composite was generated from the covalent reaction between 2-nitro-1,4-phenylenediamine (NPD) and trialdehyde phloroglucinol (Tp), and then animated by glutamate (Glu) to attach the Pd NPs. Next, the "signal on" ECL biosensor was constructed by the coordination assembly of thiolation capture DNA (cDNA) onto the Pd NPs@COFs modified electrode. After the aptamer recognition of progesterone (P4) with hairpin DNA 1 (HP1), the Exo. III cycle reaction was initiated with HP2 to generate free DNA, which hybridized with cDNA to form double-stranded DNA (dsDNA). For that, the RuP was embedded into the groove of dsDNA and achieved the ultrasensitive detection of P4 with a lower limit of detection (LOD) down to 0.45 pM, as well as the excellent selectivity and stability. This work expands the COFs-based materials application in ECL signal amplification and valuable DNA cyclic reaction in biochemical testing field.


Subject(s)
Biosensing Techniques , Electrochemical Techniques , Exodeoxyribonucleases , Metal Nanoparticles , Metal-Organic Frameworks , Palladium , Progesterone , Metal Nanoparticles/chemistry , Metal-Organic Frameworks/chemistry , Palladium/chemistry , Progesterone/analysis , Progesterone/chemistry , Biosensing Techniques/methods , Electrochemical Techniques/methods , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/metabolism , Limit of Detection , Luminescent Measurements/methods , Humans , DNA/chemistry
4.
Anal Biochem ; 691: 115547, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38670419

ABSTRACT

MicroRNAs (miRNAs) can serve as biomarkers for early-diagnosis, therapy, and postoperative care of cervical cancer. Sensitive and reliable quantification of miRNA remains a huge challenge due to its low expressing levels and background interference. Herein, we propose a novel exonuclease-III (Exo-III)-propelled DNAzyme cascade for sensitive and high-efficient miRNA analysis. This method involves the engineering of compact DNAzyme hairpin probes, including the H1 probe and H2 probe. The H1 probe is designed with exposed analyte recognition subunits that can specifically recognize target miRNA. This recognition triggers two processes: Exo-iii-assisted target regeneration and successive substrate cleavage catalyzed by DNAzyme. The unique character of Exo-III that catalyzes removal of mononucleotides from the blunt or recessed 3'-OH termini of dsDNA confers the approach with a minimal background signal. The multiple signal cycles provided an abundant signal amplification and consequently, the method exhibited a low limit of detection of 3.12 fM, and a better specificity over several homologous miRNAs. In summary, this powerful Exo-III driven DNAzyme cascaded system offers broader and more adaptable methods for comprehending the activities of miRNA in various biological occurrences.


Subject(s)
DNA, Catalytic , Exodeoxyribonucleases , MicroRNAs , Uterine Cervical Neoplasms , MicroRNAs/analysis , MicroRNAs/genetics , MicroRNAs/metabolism , DNA, Catalytic/metabolism , DNA, Catalytic/chemistry , DNA, Catalytic/genetics , Humans , Exodeoxyribonucleases/metabolism , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/genetics , Female , Limit of Detection , Biosensing Techniques/methods
5.
Nucleic Acids Res ; 52(7): 4067-4078, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38471810

ABSTRACT

Mitochondrial genome maintenance exonuclease 1 (MGME1) helps to ensure mitochondrial DNA (mtDNA) integrity by serving as an ancillary 5'-exonuclease for DNA polymerase γ. Curiously, MGME1 exhibits unique bidirectionality in vitro, being capable of degrading DNA from either the 5' or 3' end. The structural basis of this bidirectionally and, particularly, how it processes DNA from the 5' end to assist in mtDNA maintenance remain unclear. Here, we present a crystal structure of human MGME1 in complex with a 5'-overhang DNA, revealing that MGME1 functions as a rigid DNA clamp equipped with a single-strand (ss)-selective arch, allowing it to slide on single-stranded DNA in either the 5'-to-3' or 3'-to-5' direction. Using a nuclease activity assay, we have dissected the structural basis of MGME1-derived DNA cleavage patterns in which the arch serves as a ruler to determine the cleavage site. We also reveal that MGME1 displays partial DNA-unwinding ability that helps it to better resolve 5'-DNA flaps, providing insights into MGME1-mediated 5'-end processing of nascent mtDNA. Our study builds on previously solved MGME1-DNA complex structures, finally providing the comprehensive functional mechanism of this bidirectional, ss-specific exonuclease.


Subject(s)
DNA, Mitochondrial , Exodeoxyribonucleases , Genome, Mitochondrial , Humans , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , DNA, Mitochondrial/chemistry , Exodeoxyribonucleases/metabolism , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/genetics , Crystallography, X-Ray , Models, Molecular , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/chemistry , Nucleic Acid Conformation , DNA Polymerase gamma/metabolism , DNA Polymerase gamma/genetics , DNA Polymerase gamma/chemistry
6.
Bioorg Med Chem ; 102: 117660, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38442524

ABSTRACT

Werner (WRN) syndrome protein is a multifunctional enzyme with helicase, ATPase, and exonuclease activities that are necessary for numerous DNA-related transactions in the human cell. Recent studies identified WRN as a synthetic lethal target in cancers. In this study, a series of new N-arylquinazoline-4-amine analogs were designed and synthesized based on structure optimization of quinazoline. The structures of the thirty-two newly synthesized compounds were confirmed by 1H NMR, 13C NMR and ESI-MS. The anticancer activity in vitro against chronic myeloid leukemia cells (K562), non-small cell lung cancer cells (A549), human prostate cancer cells (PC3), and cervical cancer cells (HeLa) of the target compounds was evaluated. Among them, the inhibition ratio of compounds 17d, 18a, 18b, 11 and 23a against four cancer cells at 5 µM concentration were more than 50 %. The IC50 values of compounds 18a and 18b were 0.3 ± 0.01 µM and 0.05 ± 0.02 µM in K562 cells respectively, compared with HeLa and A549 cells, 18a and 18b were more sensitive to K562 cells. In addition, the PC3 cells with WRN overexpression (PC3-WRN) was constructed, 18a and 18b and 23a were more sensitive to PC3-WRN cells compared with the control group cells (PC3-NC). Then, the cell viability of the novel WRN inhibitors were further assessed by colony formation assay. Compared with PC3-NC cells, 18b and 23a had obvious inhibitory effect on PC3-WRN cell at 1000 nM. In summary, these results indicated that the compounds 18b and 23a could be WRN protein inhibitor with potent anticancer properties in vitro.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , RecQ Helicases , Exodeoxyribonucleases/metabolism , HeLa Cells
7.
Phytomedicine ; 128: 155404, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38507852

ABSTRACT

BACKGROUND: The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon (IFN) genes (STING) pathway is critical in the innate immune system and can be mobilized by cytosolic DNA. The various inflammatory and autoimmune diseases progression is highly correlated with aberrant cGAS-STING pathway activation. While some cGAS-STING pathway inhibitor were identified, there are no drugs that can be applied to the clinic. Compound Danshen Dripping Pill (CDDP) has been successfully used in clinic around the world, but the most common application is limited to cardiovascular disease. Therefore, the purpose of the present investigation was to examine whether CDDP inhibits the cGAS-STING pathway and could be used as a therapeutic agent for multiple cGAS-STING-triggered diseases. METHODS: BMDMs, THP1 cells or Trex1-/- BMDMs were stimulated with various cGAS-STING-agonists after pretreatment with CDDP to detect the function of CDDP on IFN-ß and ISGs productionn. Next, we detect the influence on IRF3 and P65 nuclear translocation, STING oligomerization and STING-TBK1-IRF3 complex formation of CDDP. Additionally, the DMXAA-mediated activation mice model of cGAS-STING pathway was used to study the effects of CDDP. Trex1-/- mice model and HFD-mediated obesity model were established to clarify the efficacy of CDDP on inflammatory and autoimmune diseases. RESULTS: CDDP efficacy suppressed the IRF3 phosphorylation or the generation of IFN-ß, ISGs, IL-6 and TNF-α. Mechanistically, CDDP did not influence the STING oligomerization and IRF3-TBK1 and STING-IRF3 interaction, but remarkably eliminated the STING-TBK1 interaction, ultimately blocking the downstream responses. In addition, we also clarified that CDDP could suppress cGAS-STING pathway activation triggered by DMXAA, in vivo. Consistently, CDDP could alleviate multi-organ inflammatory responses in Trex1-/- mice model and attenuate the inflammatory disorders, incleding obesity-induced insulin resistance. CONCLUSION: CDDP is a specifically cGAS-STING pathway inhibitor. Furthermore, we provide novel mechanism for CDDP and discovered a clinical agent for the therapy of cGAS-STING-triggered inflammatory and autoimmune diseases.


Subject(s)
Camphanes , Drugs, Chinese Herbal , Exodeoxyribonucleases , Membrane Proteins , Mice, Inbred C57BL , Nucleotidyltransferases , Panax notoginseng , Protein Serine-Threonine Kinases , Salvia miltiorrhiza , Animals , Membrane Proteins/metabolism , Salvia miltiorrhiza/chemistry , Humans , Protein Serine-Threonine Kinases/metabolism , Mice , Drugs, Chinese Herbal/pharmacology , Nucleotidyltransferases/metabolism , Exodeoxyribonucleases/metabolism , Interferon Regulatory Factor-3/metabolism , Phosphoproteins/metabolism , Signal Transduction/drug effects , THP-1 Cells , Male , Interferon-beta/metabolism , Mice, Knockout
8.
Nucleic Acids Res ; 52(7): 3722-3739, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38321948

ABSTRACT

Telomeres protect chromosome ends and are distinguished from DNA double-strand breaks (DSBs) by means of a specialized chromatin composed of DNA repeats bound by a multiprotein complex called shelterin. We investigated the role of telomere-associated proteins in establishing end-protection by studying viable mutants lacking these proteins. Mutants were studied using a Schizosaccharomyces pombe model system that induces cutting of a 'proto-telomere' bearing telomere repeats to rapidly form a new stable chromosomal end, in contrast to the rapid degradation of a control DSB. Cells lacking the telomere-associated proteins Taz1, Rap1, Poz1 or Rif1 formed a chromosome end that was stable. Surprisingly, cells lacking Ccq1, or impaired for recruiting Ccq1 to the telomere, converted the cleaved proto-telomere to a rapidly degraded DSB. Ccq1 recruits telomerase, establishes heterochromatin and affects DNA damage checkpoint activation; however, these functions were separable from protection of the new telomere by Ccq1. In cells lacking Ccq1, telomere degradation was greatly reduced by eliminating the nuclease activity of Mre11 (part of the Mre11-Rad50-Nbs1/Xrs2 DSB processing complex), and higher amounts of nuclease-deficient Mre11 associated with the new telomere. These results demonstrate a novel function for S. pombe Ccq1 to effect end-protection by restraining Mre11-dependent degradation of the DNA end.


Subject(s)
DNA Breaks, Double-Stranded , Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Telomere-Binding Proteins , Telomere , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Telomere-Binding Proteins/metabolism , Telomere-Binding Proteins/genetics , Telomere/metabolism , Telomere/genetics , Shelterin Complex/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Exodeoxyribonucleases/metabolism , Exodeoxyribonucleases/genetics , Telomerase/metabolism , Telomerase/genetics , Mutation , MRE11 Homologue Protein/metabolism , MRE11 Homologue Protein/genetics
9.
J Biol Chem ; 300(3): 105708, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38311177

ABSTRACT

A DNA double-strand break (DSB) is one of the most dangerous types of DNA damage that is repaired largely by homologous recombination or nonhomologous end-joining (NHEJ). The interplay of repair factors at the break directs which pathway is used, and a subset of these factors also function in more mutagenic alternative (alt) repair pathways. Resection is a key event in repair pathway choice and extensive resection, which is a hallmark of homologous recombination, and it is mediated by two nucleases, Exo1 and Dna2. We observed differences in resection and repair outcomes in cells harboring nuclease-dead dna2-1 compared with dna2Δ pif1-m2 that could be attributed to the level of Exo1 recovered at DSBs. Cells harboring dna2-1 showed reduced Exo1 localization, increased NHEJ, and a greater resection defect compared with cells where DNA2 was deleted. Both the resection defect and the increased rate of NHEJ in dna2-1 mutants were reversed upon deletion of KU70 or ectopic expression of Exo1. By contrast, when DNA2 was deleted, Exo1 and Ku70 recovery levels did not change; however, Nej1 increased as did the frequency of alt-end joining/microhomology-mediated end-joining repair. Our findings demonstrate that decreased Exo1 at DSBs contributed to the resection defect in cells expressing inactive Dna2 and highlight the complexity of understanding how functionally redundant factors are regulated in vivo to promote genome stability.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA Helicases , DNA-Binding Proteins , Exodeoxyribonucleases , Saccharomyces cerevisiae Proteins , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
10.
Nucleic Acids Res ; 52(8): 4328-4343, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38407383

ABSTRACT

Meiotic recombination is of central importance for the proper segregation of homologous chromosomes, but also for creating genetic diversity. It is initiated by the formation of double-strand breaks (DSBs) in DNA catalysed by evolutionarily conserved Spo11, together with additional protein partners. Difficulties in purifying the Spo11 protein have limited the characterization of its biochemical properties and of its interactions with other DSB proteins. In this study, we have purified fragments of Spo11 and show for the first time that Spo11 can physically interact with Mre11 and modulates its DNA binding, bridging, and nuclease activities. The interaction of Mre11 with Spo11 requires its far C-terminal region, which is in line with the severe meiotic phenotypes of various mre11 mutations located at the C-terminus. Moreover, calibrated ChIP for Mre11 shows that Spo11 promotes Mre11 recruitment to chromatin, independent of DSB formation. A mutant deficient in Spo11 interaction severely reduces the association of Mre11 with meiotic chromatin. Consistent with the reduction of Mre11 foci in this mutant, it strongly impedes DSB formation, leading to spore death. Our data provide evidence that physical interaction between Spo11 and Mre11, together with end-bridging, promote normal recruitment of Mre11 to hotspots and DSB formation.


Subject(s)
Chromatin , DNA Breaks, Double-Stranded , Endodeoxyribonucleases , Meiosis , Saccharomyces cerevisiae Proteins , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/genetics , Meiosis/genetics , Chromatin/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Protein Binding , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Mutation , Exodeoxyribonucleases/metabolism , Exodeoxyribonucleases/genetics
11.
Nucleic Acids Res ; 52(5): 2355-2371, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38180815

ABSTRACT

The yeast Rif2 protein is known to inhibit Mre11 nuclease and the activation of Tel1 kinase through a short motif termed MIN, which binds the Rad50 subunit and simulates its ATPase activity in vitro. The mechanism by which Rif2 restrains Tel1 activation and the consequences of this inhibition at DNA double-strand breaks (DSBs) are poorly understood. In this study, we employed AlphaFold-Multimer modelling to pinpoint and validate the interaction surface between Rif2 MIN and Rad50. We also engineered the rif2-S6E mutation that enhances the inhibitory effect of Rif2 by increasing Rif2-Rad50 interaction. Unlike rif2Δ, the rif2-S6E mutation impairs hairpin cleavage. Furthermore, it diminishes Tel1 activation by inhibiting Tel1 binding to DSBs while leaving MRX association unchanged, indicating that Rif2 can directly inhibit Tel1 recruitment to DSBs. Additionally, Rif2S6E reduces Tel1-MRX interaction and increases stimulation of ATPase by Rad50, indicating that Rif2 binding to Rad50 induces an ADP-bound MRX conformation that is not suitable for Tel1 binding. The decreased Tel1 recruitment to DSBs in rif2-S6E cells impairs DSB end-tethering and this bridging defect is suppressed by expressing a Tel1 mutant variant that increases Tel1 persistence at DSBs, suggesting a direct role for Tel1 in the bridging of DSB ends.


Subject(s)
DNA-Binding Proteins , Protein Serine-Threonine Kinases , Saccharomyces cerevisiae Proteins , Telomere-Binding Proteins , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , DNA/genetics , DNA/metabolism , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Telomere-Binding Proteins/metabolism
12.
Nucleic Acids Res ; 52(5): 2578-2589, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38261972

ABSTRACT

The loading of RecA onto ssDNA by RecBCD is an essential step of RecBCD-mediated homologous recombination. RecBCD facilitates RecA-loading onto ssDNA in a χ-dependent manner via its RecB nuclease domain (RecBn). Before recognition of χ, RecBn is sequestered through interactions with RecBCD. It was proposed that upon χ-recognition, RecBn undocks, allowing RecBn to swing out via a contiguous 70 amino acid linker to reveal the RecA-loading surface, and then recruit and load RecA onto ssDNA. We tested this hypothesis by examining the interactions between RecBn (RecB928-1180) and truncated RecBCD (RecB1-927CD) lacking the nuclease domain. The reconstituted complex of RecB1-927CD and RecBn is functional in vitro and in vivo. Our results indicate that despite being covalently severed from RecB1-927CD, RecBn can still load RecA onto ssDNA, establishing that RecBn does not function while only remaining tethered to the RecBCD complex via the linker. Instead, RecBCD undergoes a χ-induced intramolecular rearrangement to reveal the RecA-loading surface.


Subject(s)
Escherichia coli Proteins , Exodeoxyribonuclease V , Rec A Recombinases , DNA, Single-Stranded/genetics , Endonucleases/metabolism , Escherichia coli Proteins/metabolism , Exodeoxyribonuclease V/metabolism , Exodeoxyribonucleases/metabolism , Rec A Recombinases/metabolism
13.
Mol Cell ; 84(4): 659-674.e7, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38266640

ABSTRACT

Inactivating mutations in the BRCA1 and BRCA2 genes impair DNA double-strand break (DSB) repair by homologous recombination (HR), leading to chromosomal instability and cancer. Importantly, BRCA1/2 deficiency also causes therapeutically targetable vulnerabilities. Here, we identify the dependency on the end resection factor EXO1 as a key vulnerability of BRCA1-deficient cells. EXO1 deficiency generates poly(ADP-ribose)-decorated DNA lesions during S phase that associate with unresolved DSBs and genomic instability in BRCA1-deficient but not in wild-type or BRCA2-deficient cells. Our data indicate that BRCA1/EXO1 double-deficient cells accumulate DSBs due to impaired repair by single-strand annealing (SSA) on top of their HR defect. In contrast, BRCA2-deficient cells retain SSA activity in the absence of EXO1 and hence tolerate EXO1 loss. Consistent with a dependency on EXO1-mediated SSA, we find that BRCA1-mutated tumors show elevated EXO1 expression and increased SSA-associated genomic scars compared with BRCA1-proficient tumors. Overall, our findings uncover EXO1 as a promising therapeutic target for BRCA1-deficient tumors.


Subject(s)
BRCA1 Protein , Neoplasms , Humans , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , DNA Damage , DNA Repair , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Homologous Recombination
14.
Biochem Biophys Res Commun ; 695: 149464, 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38217957

ABSTRACT

DNA double strand breaks (DSBs) can be detrimental to the cell and need to be efficiently repaired. A first step in DSB repair is to bring the free ends in close proximity to enable ligation by non-homologous end-joining (NHEJ), while the more precise, but less available, repair by homologous recombination (HR) requires close proximity of a sister chromatid. The human MRE11-RAD50-NBS1 (MRN) complex, Mre11-Rad50-Xrs2 (MRX) in yeast, is involved in both repair pathways. Here we use nanofluidic channels to study, on the single DNA molecule level, how MRN, MRX and their constituents interact with long DNA and promote DNA bridging. Nanofluidics is a suitable method to study reactions on DNA ends since no anchoring of the DNA end(s) is required. We demonstrate that NBS1 and Xrs2 play important, but differing, roles in the DNA tethering by MRN and MRX. NBS1 promotes DNA bridging by MRN consistent with tethering of a repair template. MRX shows a "synapsis-like" DNA end-bridging, stimulated by the Xrs2 subunit. Our results highlight the different ways MRN and MRX bridge DNA, and the results are in agreement with their key roles in HR and NHEJ, respectively, and contribute to the understanding of the roles of NBS1 and Xrs2 in DSB repair.


Subject(s)
DNA-Binding Proteins , Endodeoxyribonucleases , Saccharomyces cerevisiae Proteins , Humans , DNA/metabolism , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
15.
Proc Natl Acad Sci U S A ; 121(5): e2319644121, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38271335

ABSTRACT

Exonuclease VII (ExoVII) is a ubiquitous bacterial nuclease. Encoded by the xseA and xseB genes, ExoVII participates in multiple nucleic acid-dependent pathways including the processing of multicopy single-stranded DNA and the repair of covalent DNA-protein crosslinks (DPCs). Although many biochemical properties of ExoVII have been defined, little is known about its structure/function relationships. Here, we use cryoelectron microscopy (cryoEM) to determine that Escherichia coli ExoVII comprises a highly elongated XseA4·XseB24 holo-complex. Each XseA subunit dimerizes through a central extended α-helical segment decorated by six XseB subunits and a C-terminal, domain-swapped ß-barrel element; two XseA2·XseB12 subcomplexes further associate using N-terminal OB (oligonucleotide/oligosaccharide-binding) folds and catalytic domains to form a spindle-shaped, catenated octaicosamer. The catalytic domains of XseA, which adopt a nuclease fold related to 3-dehydroquinate dehydratases, are sequestered in the center of the complex and accessible only through large pores formed between XseA tetramers. The architectural organization of ExoVII, combined with biochemical studies, indicate that substrate selectivity is controlled by steric access to its nuclease elements and that tetramer dissociation results from substrate DNA binding. Despite a lack of sequence and fold homology, the physical organization of ExoVII is reminiscent of Mre11·Rad50/SbcCD ATP (adenosine triphosphate)-dependent nucleases used in the repair of double-stranded DNA breaks, including those formed by DPCs through aberrant topoisomerase activity, suggesting that there may have been convergent evolutionary pressure to contend with such damage events.


Subject(s)
Escherichia coli Proteins , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Exodeoxyribonucleases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Cryoelectron Microscopy , DNA , DNA Repair
16.
J Transl Med ; 22(1): 104, 2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38279172

ABSTRACT

Prostate cancer (PCa) is one of the most common malignant tumors affecting the male genitourinary system. However, there is currently a lack of effective treatments for patients with advanced prostate cancer, which significantly impacts men's overall health. Exonuclease 1 (EXO1), a protein with mismatch repair and recombination functions, has been found to play a vital role in various diseases. In our study, we discovered that EXO1 acts as a novel biomarker of PCa, which promotes prostate cancer progression by regulating lipid metabolism reprogramming in prostate cancer cells. Mechanistically, EXO1 promotes the expression of SREBP1 by inhibiting the P53 signaling pathway. In summary, our findings suggest that EXO1 regulated intracellular lipid reprogramming through the P53/SREBP1 axis, thus promoting PCa progression. The result could potentially lead to new insights and therapeutic targets for diagnosing and treating PCa.


Subject(s)
Prostatic Neoplasms , Tumor Suppressor Protein p53 , Humans , Male , Tumor Suppressor Protein p53/metabolism , Lipid Metabolism , Prostatic Neoplasms/pathology , Lipids , Exodeoxyribonucleases/metabolism , DNA Repair Enzymes
17.
Cell Rep ; 43(1): 113637, 2024 01 23.
Article in English | MEDLINE | ID: mdl-38175749

ABSTRACT

TREX2, a 3'-5' exonuclease, is a part of the DNA damage tolerance (DDT) pathway that stabilizes replication forks (RFs) by ubiquitinating PCNA along with the ubiquitin E3 ligase RAD18 and other DDT factors. Mismatch repair (MMR) corrects DNA polymerase errors, including base mismatches and slippage. Here we demonstrate that TREX2 deletion reduces mutations in cells upon exposure to genotoxins, including those that cause base lesions and DNA polymerase slippage. Importantly, we show that TREX2 generates most of the spontaneous mutations in MMR-mutant cells derived from mice and people. TREX2-induced mutagenesis is dependent on the nuclease and DNA-binding attributes of TREX2. RAD18 deletion also reduces spontaneous mutations in MMR-mutant cells, albeit to a lesser degree. Inactivation of both MMR and TREX2 additively increases RF stalls, while it decreases DNA breaks, consistent with a synthetic phenotype.


Subject(s)
DNA-Directed DNA Polymerase , Mutagens , Humans , Mice , Animals , Mutagenesis , DNA-Directed DNA Polymerase/metabolism , Mutation , Ubiquitin/metabolism , DNA Replication , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Phosphoproteins/genetics , DNA-Binding Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
18.
Int J Biol Macromol ; 255: 128305, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37992942

ABSTRACT

Leukemia is a type of malignant hematological disease that is generally resistant to chemotherapy and has poor therapeutic outcomes. Werner (WRN) DNA helicase, an important member of the RecQ family of helicases, plays an important role in DNA repair and telomere stability maintenance. WRN gene dysfunction leads to premature aging and predisposes humans to various types of cancers. However, the biological function of WRN in cancer remains unknown. In this study, the expression of this RecQ family helicase was investigated in different types of leukemia cells, and the leukemia cell line K562 with high WRN expression was selected to construct a WRN knockdown cell line. The results showed that WRN knockdown inhibited leukemia occurrence and development by regulating the proliferation, cell cycle, differentiation, and aging of cells and other biological processes. The results of transcriptome sequencing revealed that WRN promoted the sensitivity of leukemia cells to the DNA damage inducer Etoposide by regulating cell cycle-related proteins, such as CDC2, cyclin B1, p16, and p21, as well as key proteins in DNA damage repair pathways, such as p53, RAD50, RAD51, and MER11. Our findings show that WRN helicase is a promising potential target for leukemia treatment, providing new ideas for the development of targeted drugs against leukemia.


Subject(s)
Exodeoxyribonucleases , Leukemia , Humans , Werner Syndrome Helicase/genetics , Werner Syndrome Helicase/metabolism , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , RecQ Helicases/genetics , RecQ Helicases/metabolism , Cell Cycle/genetics , DNA Repair , DNA Damage , Leukemia/genetics
19.
Genes Dev ; 37(19-20): 913-928, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37932011

ABSTRACT

Addiction to the WRN helicase is a unique vulnerability of human cancers with high levels of microsatellite instability (MSI-H). However, while prolonged loss of WRN ultimately leads to cell death, little is known about how MSI-H cancers initially respond to acute loss of WRN-knowledge that would be helpful for informing clinical development of WRN targeting therapy, predicting possible resistance mechanisms, and identifying useful biomarkers of successful WRN inhibition. Here, we report the construction of an inducible ligand-mediated degradation system in which the stability of endogenous WRN protein can be rapidly and specifically tuned, enabling us to track the complete sequence of cellular events elicited by acute loss of WRN function. We found that WRN degradation leads to immediate accrual of DNA damage in a replication-dependent manner that curiously did not robustly engage checkpoint mechanisms to halt DNA synthesis. As a result, WRN-degraded MSI-H cancer cells accumulate DNA damage across multiple replicative cycles and undergo successive rounds of increasingly aberrant mitoses, ultimately triggering cell death. Of potential therapeutic importance, we found no evidence of any generalized mechanism by which MSI-H cancers could adapt to near-complete loss of WRN. However, under conditions of partial WRN degradation, addition of low-dose ATR inhibitor significantly increased their combined efficacy to levels approaching full inactivation of WRN. Overall, our results provide the first comprehensive view of molecular events linking upstream inhibition of WRN to subsequent cell death and suggest that dual targeting of WRN and ATR might be a useful strategy for treating MSI-H cancers.


Subject(s)
DNA Replication , Neoplasms , Humans , DNA Replication/genetics , DNA Helicases/metabolism , Microsatellite Repeats , DNA Damage , Neoplasms/drug therapy , Neoplasms/genetics , RecQ Helicases/genetics , RecQ Helicases/metabolism , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Werner Syndrome Helicase/genetics , Werner Syndrome Helicase/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism
20.
Nat Commun ; 14(1): 6265, 2023 10 07.
Article in English | MEDLINE | ID: mdl-37805499

ABSTRACT

Accumulation of single stranded DNA (ssDNA) gaps in the nascent strand during DNA replication has been associated with cytotoxicity and hypersensitivity to genotoxic stress, particularly upon inactivation of the BRCA tumor suppressor pathway. However, how ssDNA gaps contribute to genotoxicity is not well understood. Here, we describe a multi-step nucleolytic processing of replication stress-induced ssDNA gaps which converts them into cytotoxic double stranded DNA breaks (DSBs). We show that ssDNA gaps are extended bidirectionally by MRE11 in the 3'-5' direction and by EXO1 in the 5'-3' direction, in a process which is suppressed by the BRCA pathway. Subsequently, the parental strand at the ssDNA gap is cleaved by the MRE11 endonuclease generating a double strand break. We also show that exposure to bisphenol A (BPA) and diethylhexyl phthalate (DEHP), which are widespread environmental contaminants due to their use in plastics manufacturing, causes nascent strand ssDNA gaps during replication. These gaps are processed through the same mechanism described above to generate DSBs. Our work sheds light on both the relevance of ssDNA gaps as major determinants of genomic instability, as well as the mechanism through which they are processed to generate genomic instability and cytotoxicity.


Subject(s)
DNA Repair , Exodeoxyribonucleases , Humans , Exodeoxyribonucleases/metabolism , Endonucleases/genetics , Endonucleases/metabolism , DNA/genetics , Genomic Instability , DNA Replication , DNA, Single-Stranded/genetics , DNA Repair Enzymes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...