Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 163
Filter
1.
Nat Commun ; 15(1): 5789, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987539

ABSTRACT

The outcome of CRISPR-Cas-mediated genome modifications is dependent on DNA double-strand break (DSB) processing and repair pathway choice. Homology-directed repair (HDR) of protein-blocked DSBs requires DNA end resection that is initiated by the endonuclease activity of the MRE11 complex. Using reconstituted reactions, we show that Cas9 breaks are unexpectedly not directly resectable by the MRE11 complex. In contrast, breaks catalyzed by Cas12a are readily processed. Cas9, unlike Cas12a, bridges the broken ends, preventing DSB detection and processing by MRE11. We demonstrate that Cas9 must be dislocated after DNA cleavage to allow DNA end resection and repair. Using single molecule and bulk biochemical assays, we next find that the HLTF translocase directly removes Cas9 from broken ends, which allows DSB processing by DNA end resection or non-homologous end-joining machineries. Mechanistically, the activity of HLTF requires its HIRAN domain and the release of the 3'-end generated by the cleavage of the non-target DNA strand by the Cas9 RuvC domain. Consequently, HLTF removes the H840A but not the D10A Cas9 nickase. The removal of Cas9 H840A by HLTF explains the different cellular impact of the two Cas9 nickase variants in human cells, with potential implications for gene editing.


Subject(s)
CRISPR-Associated Protein 9 , CRISPR-Cas Systems , DNA Breaks, Double-Stranded , DNA , Humans , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Protein 9/genetics , DNA/metabolism , DNA/genetics , MRE11 Homologue Protein/metabolism , MRE11 Homologue Protein/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/genetics , Gene Editing , Endonucleases/metabolism , Endonucleases/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/genetics , DNA End-Joining Repair , DNA Cleavage , Transcription Factors/metabolism , Transcription Factors/genetics
2.
Mol Cell ; 84(12): 2223-2237.e4, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38870937

ABSTRACT

In Saccharomyces cerevisiae (S. cerevisiae), Mre11-Rad50-Xrs2 (MRX)-Sae2 nuclease activity is required for the resection of DNA breaks with secondary structures or protein blocks, while in humans, the MRE11-RAD50-NBS1 (MRN) homolog with CtIP is needed to initiate DNA end resection of all breaks. Phosphorylated Sae2/CtIP stimulates the endonuclease activity of MRX/N. Structural insights into the activation of the Mre11 nuclease are available only for organisms lacking Sae2/CtIP, so little is known about how Sae2/CtIP activates the nuclease ensemble. Here, we uncover the mechanism of Mre11 activation by Sae2 using a combination of AlphaFold2 structural modeling of biochemical and genetic assays. We show that Sae2 stabilizes the Mre11 nuclease in a conformation poised to cleave substrate DNA. Several designs of compensatory mutations establish how Sae2 activates MRX in vitro and in vivo, supporting the structural model. Finally, our study uncovers how human CtIP, despite considerable sequence divergence, employs a similar mechanism to activate MRN.


Subject(s)
DNA-Binding Proteins , Endodeoxyribonucleases , Endonucleases , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/metabolism , Endonucleases/metabolism , Endonucleases/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/chemistry , Humans , Exodeoxyribonucleases/metabolism , Exodeoxyribonucleases/genetics , Models, Molecular , Phosphorylation , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , DNA Breaks, Double-Stranded , Acid Anhydride Hydrolases/metabolism , Acid Anhydride Hydrolases/genetics , Mutation , MRE11 Homologue Protein/metabolism , MRE11 Homologue Protein/genetics , DNA Repair , Enzyme Activation
3.
Nat Commun ; 15(1): 5423, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926338

ABSTRACT

Oncogene-induced senescence (OIS) arrests cell proliferation in response to replication stress (RS) induced by oncogenes. OIS depends on the DNA damage response (DDR), but also on the cGAS-STING pathway, which detects cytosolic DNA and induces type I interferons (IFNs). Whether and how RS and IFN responses cooperate to promote OIS remains unknown. Here, we show that the induction of OIS by the H-RASV12 oncogene in immortalized human fibroblasts depends on the MRE11 nuclease. Indeed, treatment with the MRE11 inhibitor Mirin prevented RS, micronuclei formation and IFN response induced by RASV12. Overexpression of the cytosolic nuclease TREX1 also prevented OIS. Conversely, overexpression of a dominant negative mutant of TREX1 or treatment with IFN-ß was sufficient to induce RS and DNA damage, independent of RASV12 induction. These data suggest that the IFN response acts as a positive feedback loop to amplify DDR in OIS through a process regulated by MRE11 and TREX1.


Subject(s)
Cellular Senescence , DNA Damage , DNA Replication , Exodeoxyribonucleases , MRE11 Homologue Protein , Phosphoproteins , Signal Transduction , Humans , Exodeoxyribonucleases/metabolism , Exodeoxyribonucleases/genetics , Phosphoproteins/metabolism , Phosphoproteins/genetics , MRE11 Homologue Protein/metabolism , MRE11 Homologue Protein/genetics , Cellular Senescence/genetics , Fibroblasts/metabolism , Interferon-beta/metabolism , Interferon-beta/genetics
4.
J Clin Invest ; 134(13)2024 May 14.
Article in English | MEDLINE | ID: mdl-38758740

ABSTRACT

The diversity of structural variants (SVs) in melanoma and how they impact oncogenesis are incompletely known. We performed harmonized analysis of SVs across melanoma histologic and genomic subtypes, and we identified distinct global properties between subtypes. These included the frequency and size of SVs and SV classes, their relation to chromothripsis events, and the impact on cancer-related genes of SVs that alter topologically associated domain (TAD) boundaries. Following our prior identification of double-stranded break repair deficiency in a subset of triple-wild-type cutaneous melanoma, we identified MRE11 and NBN loss-of-function SVs in melanomas with this mutational signature. Experimental knockouts of MRE11 and NBN, followed by olaparib cell viability assays in melanoma cells, indicated that dysregulation of each of these genes may cause sensitivity to PARP inhibitors in cutaneous melanomas. Broadly, harmonized analysis of melanoma SVs revealed distinct global genomic properties and molecular drivers, which may have biological and therapeutic impact.


Subject(s)
Melanoma , Melanoma/genetics , Melanoma/pathology , Melanoma/metabolism , Humans , Cell Line, Tumor , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/metabolism , Carcinogenesis/genetics , MRE11 Homologue Protein/genetics , MRE11 Homologue Protein/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phthalazines/pharmacology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Genomic Structural Variation/genetics , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
5.
Cell Rep ; 43(4): 114024, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38581679

ABSTRACT

Mouse embryonic stem cells (mESCs) in the primed pluripotency state, which resembles the post-implantation epiblast, can be de-differentiated in culture to a naive state that resembles the pre-implantation inner cell mass. We report that primed-to-naive mESC transition entails a significant slowdown of DNA replication forks and the compensatory activation of dormant origins. Using isolation of proteins on nascent DNA coupled to mass spectrometry, we identify key changes in replisome composition that are responsible for these effects. Naive mESC forks are enriched in MRE11 nuclease and other DNA repair proteins. MRE11 is recruited to newly synthesized DNA in response to transcription-replication conflicts, and its inhibition or genetic downregulation in naive mESCs is sufficient to restore the fork rate of primed cells. Transcriptomic analyses indicate that MRE11 exonuclease activity is required for the complete primed-to-naive mESC transition, demonstrating a direct link between DNA replication dynamics and the mESC de-differentiation process.


Subject(s)
DNA Replication , MRE11 Homologue Protein , Animals , Mice , MRE11 Homologue Protein/metabolism , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/cytology , Cell Dedifferentiation , DNA-Binding Proteins/metabolism
6.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 46(2): 232-241, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-38686720

ABSTRACT

DNA is susceptible to various factors in vitro and in vivo and experience different forms of damage,among which double-strand break(DSB)is a deleterious form.To maintain the stability of genetic information,organisms have developed multiple mechanisms to repair DNA damage.Among these mechanisms,homologous recombination(HR)is praised for the high accuracy.The MRE11-RAD50-NBS1(MRN)complex plays an important role in HR and is conserved across different species.The knowledge on the MRN complex mainly came from the previous studies in Saccharomyces cerevisiae and Caenorhabditis elegans,while studies in the last decades have revealed the role of mammalian MRN complex in DNA repair of higher animals.In this review,we first introduces the MRN complex regarding the composition,structure,and roles in HR.In addition,we discuss the human diseases such as ataxia-telangiectasia-like disorder,Nijmegen breakage syndrome,and Nijmegen breakage syndrome-like disorder that are caused by dysfunctions in the MRN complex.Furthermore,we summarize the mouse models established to study the clinical phenotypes of the above diseases.


Subject(s)
Acid Anhydride Hydrolases , Cell Cycle Proteins , DNA Repair Enzymes , DNA-Binding Proteins , MRE11 Homologue Protein , Nuclear Proteins , Humans , Acid Anhydride Hydrolases/metabolism , Acid Anhydride Hydrolases/genetics , MRE11 Homologue Protein/metabolism , MRE11 Homologue Protein/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Animals , DNA Repair , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/metabolism , Nijmegen Breakage Syndrome/metabolism , Nijmegen Breakage Syndrome/genetics
7.
Cell Mol Biol (Noisy-le-grand) ; 70(3): 219-224, 2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38650130

ABSTRACT

Mitochondrial DNA damage in retinal ganglion cells (RGCs) may be closely related to lesions of glaucoma. RGCs were cultured with different concentrations of glucose and grouped into 3 groups, namely normal control (NC) group, Low-Glu group, and High-Glu group. Cell viability was measured with cell counting kit-8, and cell apoptosis was measured using flow cytometry. The DNA damage was measured with comet assay, and the morphological changes of damaged mitochondria in RGCs were observed using TEM. Western blot analyzed the expression of MRE11, RAD50, and NBS1 protein. Cell viability of RGCs in Low-Glu and High-Glu groups were lower than that of NC group in 48 and 96 h. The cell apoptosis in NC group was 4.9%, the Low-Glu group was 12.2% and High-Glu group was 24.4%. The comet imaging showed that NC cells did not have tailings, but the low-Glu and high-Glu group cells had tailings, indicating that the DNA of RGCs had been damaged. TEM, mitochondrial membrane potential, ROS, mitochondrial oxygen consumption, and ATP content detection results showed that RGCs cultured with high glucose occurred mitochondrial morphology changes and dysfunction. MRE11, RAD50, and NBS1 protein expression associated with DNA damage repair pathway in High-Glu group declined compared with Low-Glu group. Mitochondrial DNA damage caused by high glucose will result in apoptosis of retinal ganglion cells in glaucoma.


Subject(s)
Apoptosis , Cell Survival , DNA Damage , DNA, Mitochondrial , Glucose , Membrane Potential, Mitochondrial , Reactive Oxygen Species , Retinal Ganglion Cells , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Glucose/toxicity , Glucose/pharmacology , DNA, Mitochondrial/metabolism , DNA, Mitochondrial/genetics , Apoptosis/drug effects , Cell Survival/drug effects , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Adenosine Triphosphate/metabolism , MRE11 Homologue Protein/metabolism , MRE11 Homologue Protein/genetics , Mitochondria/metabolism , Mitochondria/drug effects , Acid Anhydride Hydrolases/genetics , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , Humans , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Comet Assay , Animals
8.
Nat Commun ; 15(1): 2132, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38459011

ABSTRACT

Growth factor receptor-bound protein 2 (GRB2) is a cytoplasmic adapter for tyrosine kinase signaling and a nuclear adapter for homology-directed-DNA repair. Here we find nuclear GRB2 protects DNA at stalled replication forks from MRE11-mediated degradation in the BRCA2 replication fork protection axis. Mechanistically, GRB2 binds and inhibits RAD51 ATPase activity to stabilize RAD51 on stalled replication forks. In GRB2-depleted cells, PARP inhibitor (PARPi) treatment releases DNA fragments from stalled forks into the cytoplasm that activate the cGAS-STING pathway to trigger pro-inflammatory cytokine production. Moreover in a syngeneic mouse metastatic ovarian cancer model, GRB2 depletion in the context of PARPi treatment reduced tumor burden and enabled high survival consistent with immune suppression of cancer growth. Collective findings unveil GRB2 function and mechanism for fork protection in the BRCA2-RAD51-MRE11 axis and suggest GRB2 as a potential therapeutic target and an enabling predictive biomarker for patient selection for PARPi and immunotherapy combination.


Subject(s)
DNA Replication , Neoplasms , Animals , Humans , Mice , DNA , Genomic Instability , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , Immunity, Innate , MRE11 Homologue Protein/metabolism , Neoplasms/genetics , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism
10.
Nucleic Acids Res ; 52(6): 3146-3163, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38349040

ABSTRACT

Sensing and processing of DNA double-strand breaks (DSBs) are vital to genome stability. DSBs are primarily detected by the ATM checkpoint pathway, where the Mre11-Rad50-Nbs1 (MRN) complex serves as the DSB sensor. Subsequent DSB end resection activates the ATR checkpoint pathway, where replication protein A, MRN, and the Rad9-Hus1-Rad1 (9-1-1) clamp serve as the DNA structure sensors. ATR activation depends also on Topbp1, which is loaded onto DNA through multiple mechanisms. While different DNA structures elicit specific ATR-activation subpathways, the regulation and mechanisms of the ATR-activation subpathways are not fully understood. Using DNA substrates that mimic extensively resected DSBs, we show here that MRN and 9-1-1 redundantly stimulate Dna2-dependent long-range end resection and ATR activation in Xenopus egg extracts. MRN serves as the loading platform for ATM, which, in turn, stimulates Dna2- and Topbp1-loading. Nevertheless, MRN promotes Dna2-mediated end processing largely independently of ATM. 9-1-1 is dispensable for bulk Dna2 loading, and Topbp1 loading is interdependent with 9-1-1. ATR facilitates Mre11 phosphorylation and ATM dissociation. These data uncover that long-range end resection activates two redundant pathways that facilitate ATR checkpoint signaling and DNA processing in a vertebrate system.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , DNA Breaks, Double-Stranded , DNA Repair Enzymes , Xenopus Proteins , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA/genetics , DNA/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , MRE11 Homologue Protein/genetics , MRE11 Homologue Protein/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Xenopus laevis/genetics , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Enzyme Activation/genetics , Phosphorylation/genetics
11.
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
12.
EMBO J ; 43(6): 1043-1064, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38360996

ABSTRACT

Eukaryotic cells rely on several mechanisms to ensure that the genome is duplicated precisely once in each cell division cycle, preventing DNA over-replication and genomic instability. Most of these mechanisms limit the activity of origin licensing proteins to prevent the reactivation of origins that have already been used. Here, we have investigated whether additional controls restrict the extension of re-replicated DNA in the event of origin re-activation. In a genetic screening in cells forced to re-activate origins, we found that re-replication is limited by RAD51 and enhanced by FBH1, a RAD51 antagonist. In the presence of chromatin-bound RAD51, forks stemming from re-fired origins are slowed down, leading to frequent events of fork reversal. Eventual re-initiation of DNA synthesis mediated by PRIMPOL creates ssDNA gaps that facilitate the partial elimination of re-duplicated DNA by MRE11 exonuclease. In the absence of RAD51, these controls are abrogated and re-replication forks progress much longer than in normal conditions. Our study uncovers a safeguard mechanism to protect genome stability in the event of origin reactivation.


Subject(s)
DNA-Binding Proteins , Rad51 Recombinase , DNA/genetics , DNA Replication , DNA-Binding Proteins/metabolism , MRE11 Homologue Protein/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Humans
13.
Nature ; 625(7995): 585-592, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38200309

ABSTRACT

Oncogene-induced replication stress generates endogenous DNA damage that activates cGAS-STING-mediated signalling and tumour suppression1-3. However, the precise mechanism of cGAS activation by endogenous DNA damage remains enigmatic, particularly given that high-affinity histone acidic patch (AP) binding constitutively inhibits cGAS by sterically hindering its activation by double-stranded DNA (dsDNA)4-10. Here we report that the DNA double-strand break sensor MRE11 suppresses mammary tumorigenesis through a pivotal role in regulating cGAS activation. We demonstrate that binding of the MRE11-RAD50-NBN complex to nucleosome fragments is necessary to displace cGAS from acidic-patch-mediated sequestration, which enables its mobilization and activation by dsDNA. MRE11 is therefore essential for cGAS activation in response to oncogenic stress, cytosolic dsDNA and ionizing radiation. Furthermore, MRE11-dependent cGAS activation promotes ZBP1-RIPK3-MLKL-mediated necroptosis, which is essential to suppress oncogenic proliferation and breast tumorigenesis. Notably, downregulation of ZBP1 in human triple-negative breast cancer is associated with increased genome instability, immune suppression and poor patient prognosis. These findings establish MRE11 as a crucial mediator that links DNA damage and cGAS activation, resulting in tumour suppression through ZBP1-dependent necroptosis.


Subject(s)
Cell Transformation, Neoplastic , MRE11 Homologue Protein , Nucleosomes , Nucleotidyltransferases , Humans , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , DNA Damage , MRE11 Homologue Protein/metabolism , Necroptosis , Nucleosomes/metabolism , Nucleotidyltransferases/metabolism , Radiation, Ionizing , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Genomic Instability
14.
Biochim Biophys Acta Mol Cell Res ; 1871(2): 119654, 2024 02.
Article in English | MEDLINE | ID: mdl-38123020

ABSTRACT

The genome is frequently targeted by genotoxic agents, resulting in the formation of DNA scars. However, cells employ diverse repair mechanisms to restore DNA integrity. Among these processes, the Mre11-Rad50-Nbs1 complex detects double-strand breaks (DSBs) and recruits DNA damage response proteins such as ataxia-telangiectasia-mutated (ATM) kinase to DNA damage sites. ATM phosphorylates the transactivation domain (TAD) of the p53 tumor suppressor, which in turn regulates DNA repair, growth arrest, apoptosis, and senescence following DNA damage. The disordered glycine-arginine-rich (GAR) domain of double-strand break protein MRE11 (MRE11GAR) and its methylation are important for DSB repair, and localization to Promyelocytic leukemia nuclear bodies (PML-NBs). There is preliminary evidence that p53, PML protein, and MRE11 might co-localize and interact at DSB sites. To uncover the molecular details of these interactions, we aimed to identify the domains mediating the p53-MRE11 interaction and to elucidate the regulation of the p53-MRE11 interaction by post-translational modifications (PTMs) through a combination of biophysical techniques. We discovered that, in vitro, p53 binds directly to MRE11GAR mainly through p53TAD2 and that phosphorylation further enhances this interaction. Furthermore, we found that MRE11GAR methylation still allows for binding to p53. Overall, we demonstrated that p53 and MRE11 interaction is facilitated by disordered regions. We provide for the first time insight into the molecular details of the p53-MRE11 complex formation and elucidate potential regulatory mechanisms that will promote our understanding of the DNA damage response. Our findings suggest that PTMs regulate the p53-MRE11 interaction and subsequently their colocalization to PML-NBs upon DNA damage.


Subject(s)
Cell Cycle Proteins , Tumor Suppressor Protein p53 , Tumor Suppressor Protein p53/genetics , Cell Cycle Proteins/metabolism , MRE11 Homologue Protein/genetics , MRE11 Homologue Protein/metabolism , DNA-Binding Proteins/metabolism , DNA
15.
Cell ; 187(2): 294-311.e21, 2024 01 18.
Article in English | MEDLINE | ID: mdl-38128537

ABSTRACT

Lactylation is a lactate-induced post-translational modification best known for its roles in epigenetic regulation. Herein, we demonstrate that MRE11, a crucial homologous recombination (HR) protein, is lactylated at K673 by the CBP acetyltransferase in response to DNA damage and dependent on ATM phosphorylation of the latter. MRE11 lactylation promotes its binding to DNA, facilitating DNA end resection and HR. Inhibition of CBP or LDH downregulated MRE11 lactylation, impaired HR, and enhanced chemosensitivity of tumor cells in patient-derived xenograft and organoid models. A cell-penetrating peptide that specifically blocks MRE11 lactylation inhibited HR and sensitized cancer cells to cisplatin and PARPi. These findings unveil lactylation as a key regulator of HR, providing fresh insights into the ways in which cellular metabolism is linked to DSB repair. They also imply that the Warburg effect can confer chemoresistance through enhancing HR and suggest a potential therapeutic strategy of targeting MRE11 lactylation to mitigate the effects.


Subject(s)
DNA-Binding Proteins , MRE11 Homologue Protein , Recombinational DNA Repair , Humans , DNA , DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Homologous Recombination , MRE11 Homologue Protein/metabolism , Lactic Acid/metabolism
16.
J Clin Immunol ; 43(8): 2136-2145, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37794136

ABSTRACT

PURPOSE: The MRE11-RAD50-NBN (MRN) complex plays a key role in recognizing and signaling DNA double-strand breaks. Pathogenic variants in NBN and MRE11 give rise to the autosomal-recessive diseases, Nijmegen breakage syndrome (NBS) and ataxia telangiectasia-like disorder, respectively. The clinical consequences of pathogenic variants in RAD50 are incompletely understood. We aimed to characterize a newly identified RAD50 deficiency/NBS-like disorder (NBSLD) patient with bone marrow failure and immunodeficiency. METHODS: We report on a girl with microcephaly, mental retardation, bird-like face, short stature, bone marrow failure and B-cell immunodeficiency. We searched for candidate gene by whole-exome sequencing and analyzed the cellular phenotype of patient-derived fibroblasts using immunoblotting, radiation sensitivity assays and lentiviral complementation experiments. RESULTS: Compound heterozygosity for two variants in the RAD50 gene (p.Arg83His and p.Glu485Ter) was identified in this patient. The expression of RAD50 protein and MRN complex formation was maintained in the cells derived from this patient. DNA damage-induced activation of the ATM kinase was markedly decreased, which was restored by the expression of wild-type (WT) RAD50. Radiosensitivity appeared inconspicuous in the patient-derived cell line as assessed by colony formation assay. The RAD50R83H missense substitution did not rescue the mitotic defect in complementation experiments using RAD50-deficient fibroblasts, whereas RAD50WT did. The RAD50E485X nonsense variant was associated with in-frame skipping of exon 10 (p.Glu485_545del). CONCLUSION: These findings indicate important roles of RAD50 in human bone marrow and immune cells. RAD50 deficiency/NBSLD can manifest as a distinct inborn error of immunity characterized by bone marrow failure and B-cell immunodeficiency.


Subject(s)
Immunologic Deficiency Syndromes , Nijmegen Breakage Syndrome , Female , Humans , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , MRE11 Homologue Protein/genetics , MRE11 Homologue Protein/metabolism , Nijmegen Breakage Syndrome/genetics , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/genetics , Bone Marrow Failure Disorders
17.
Protein Sci ; 32(10): e4782, 2023 10.
Article in English | MEDLINE | ID: mdl-37705456

ABSTRACT

The repair of double-strand DNA breaks (DSBs) by homologous recombination is crucial in the maintenance of genome integrity. While the key role of the Mre11-Rad50-Nbs1 (MRN) complex in repair is well known, hSSB1 (SOSSB and OBFC2B), one of the main components of the sensor of single-stranded DNA (SOSS) protein complex, has also been shown to rapidly localize to DSB breaks and promote repair. We have previously demonstrated that hSSB1 binds directly to Nbs1, a component of the MRN complex, in a DNA damage-independent manner. However, recruitment of the MRN complex has also been demonstrated by an interaction between Integrator Complex Subunit 3 (INTS3; also known as SOSSA), another member of the SOSS complex, and Nbs1. In this study, we utilize a combined approach of in silico, biochemical, and functional experiments to uncover the molecular details of INTS3 binding to Nbs1. We demonstrate that the forkhead-associated domain of Nbs1 interacts with INTS3 via phosphorylation-dependent binding to INTS3 at Threonine 592, with contributions from Serine 590. Based on these data, we propose a model of MRN recruitment to a DSB via INTS3.


Subject(s)
Cell Cycle Proteins , Nuclear Proteins , Phosphorylation , MRE11 Homologue Protein/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Repair
18.
Int J Mol Sci ; 24(13)2023 Jun 30.
Article in English | MEDLINE | ID: mdl-37446144

ABSTRACT

The MRE11 nuclease is essential during DNA damage recognition, homologous recombination, and replication. BRCA2 plays important roles during homologous recombination and replication. Here, we show that effecting an MRE11 blockade using a prototypical inhibitor (Mirin) induces synthetic lethality (SL) in BRCA2-deficient ovarian cancer cells, HeLa cells, and 3D spheroids compared to BRCA2-proficient controls. Increased cytotoxicity was associated with double-strand break accumulation, S-phase cell cycle arrest, and increased apoptosis. An in silico analysis revealed Mirin docking onto the active site of MRE11. While Mirin sensitises DT40 MRE11+/- cells to the Top1 poison SN-38, it does not sensitise nuclease-dead MRE11 cells to this compound confirming that Mirin specifically inhibits Mre11 nuclease activity. MRE11 knockdown reduced cell viability in BRCA2-deficient PEO1 cells but not in BRCA2-proficient PEO4 cells. In a Mirin-resistant model, we show the downregulation of 53BP1 and DNA repair upregulation, leading to resistance, including in in vivo xenograft models. In a clinical cohort of human ovarian tumours, low levels of BRCA2 expression with high levels of MRE11 co-expression were linked with worse progression-free survival (PFS) (p = 0.005) and overall survival (OS) (p = 0.001). We conclude that MRE11 is an attractive SL target, and the pharmaceutical development of MRE11 inhibitors for precision oncology therapeutics may be of clinical benefit.


Subject(s)
DNA-Binding Proteins , Ovarian Neoplasms , Humans , Female , DNA-Binding Proteins/metabolism , MRE11 Homologue Protein/genetics , MRE11 Homologue Protein/metabolism , HeLa Cells , Precision Medicine , BRCA2 Protein/metabolism , DNA Repair , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Cell Line, Tumor
19.
J Virol ; 97(5): e0046123, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37098896

ABSTRACT

Parvoviruses are single-stranded DNA viruses that utilize host proteins to vigorously replicate in the nuclei of host cells, leading to cell cycle arrest. The autonomous parvovirus, minute virus of mice (MVM), forms viral replication centers in the nucleus which are adjacent to cellular DNA damage response (DDR) sites, many of which are fragile genomic regions prone to undergoing DDR during the S phase. Since the cellular DDR machinery has evolved to transcriptionally suppress the host epigenome to maintain genomic fidelity, the successful expression and replication of MVM genomes at these cellular sites suggest that MVM interacts with DDR machinery distinctly. Here, we show that efficient replication of MVM requires binding of the host DNA repair protein MRE11 in a manner that is independent of the MRE11-RAD50-NBS1 (MRN) complex. MRE11 binds to the replicating MVM genome at the P4 promoter, remaining distinct from RAD50 and NBS1, which associate with cellular DNA break sites to generate DDR signals in the host genome. Ectopic expression of wild-type MRE11 in CRISPR knockout cells rescues virus replication, revealing a dependence on MRE11 for efficient MVM replication. Our findings suggest a new model utilized by autonomous parvoviruses to usurp local DDR proteins that are crucial for viral pathogenesis and distinct from those of dependoparvoviruses, like adeno-associated virus (AAV), which require a coinfected helper virus to inactivate the local host DDR. IMPORTANCE The cellular DNA damage response (DDR) machinery protects the host genome from the deleterious consequences of DNA breaks and recognizes invading viral pathogens. DNA viruses that replicate in the nucleus have evolved distinct strategies to evade or usurp these DDR proteins. We have discovered that the autonomous parvovirus, MVM, which is used to target cancer cells as an oncolytic agent, depends on the initial DDR sensor protein MRE11 to express and replicate efficiently in host cells. Our studies reveal that the host DDR interacts with replicating MVM molecules in ways that are distinct from viral genomes being recognized as simple broken DNA molecules. These findings suggest that autonomous parvoviruses have evolved distinct mechanisms to usurp DDR proteins, which can be used to design potent DDR-dependent oncolytic agents.


Subject(s)
MRE11 Homologue Protein , Minute Virus of Mice , Parvoviridae Infections , Animals , Mice , Cell Cycle Proteins/metabolism , Discoidin Domain Receptors/genetics , Discoidin Domain Receptors/metabolism , DNA Damage , DNA Replication , Minute Virus of Mice/genetics , Parvoviridae Infections/genetics , Virus Replication/physiology , MRE11 Homologue Protein/metabolism
20.
Int J Mol Sci ; 24(6)2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36982687

ABSTRACT

The MRE11, RAD50, and NBN genes encode for the nuclear MRN protein complex, which senses the DNA double strand breaks and initiates the DNA repair. The MRN complex also participates in the activation of ATM kinase, which coordinates DNA repair with the p53-dependent cell cycle checkpoint arrest. Carriers of homozygous germline pathogenic variants in the MRN complex genes or compound heterozygotes develop phenotypically distinct rare autosomal recessive syndromes characterized by chromosomal instability and neurological symptoms. Heterozygous germline alterations in the MRN complex genes have been associated with a poorly-specified predisposition to various cancer types. Somatic alterations in the MRN complex genes may represent valuable predictive and prognostic biomarkers in cancer patients. MRN complex genes have been targeted in several next-generation sequencing panels for cancer and neurological disorders, but interpretation of the identified alterations is challenging due to the complexity of MRN complex function in the DNA damage response. In this review, we outline the structural characteristics of the MRE11, RAD50 and NBN proteins, the assembly and functions of the MRN complex from the perspective of clinical interpretation of germline and somatic alterations in the MRE11, RAD50 and NBN genes.


Subject(s)
Cell Cycle Proteins , Tumor Suppressor Proteins , Humans , Cell Cycle Proteins/metabolism , Tumor Suppressor Proteins/genetics , MRE11 Homologue Protein/genetics , MRE11 Homologue Protein/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Repair/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Acid Anhydride Hydrolases/genetics , Acid Anhydride Hydrolases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...