Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Hum Mol Genet ; 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38888340

ABSTRACT

The MRE11/RAD50/NBS1 (MRN) complex plays critical roles in cellular responses to DNA double-strand breaks. MRN is involved in end binding and processing, and it also induces cell cycle checkpoints by activating the ataxia-telangiectasia mutated (ATM) protein kinase. Hypomorphic pathogenic variants in the MRE11, RAD50, or NBS1 genes cause autosomal recessive genome instability syndromes featuring variable degrees of dwarfism, neurological defects, anemia, and cancer predisposition. Disease-associated MRN alleles include missense and nonsense variants, and many cause reduced protein levels of the entire MRN complex. However, the dramatic variability in the disease manifestation of MRN pathogenic variants is not understood. We sought to determine if low protein levels are a significant contributor to disease sequelae and therefore generated a transgenic murine model expressing MRE11 at low levels. These mice display dramatic phenotypes including small body size, severe anemia, and impaired DNA repair. We demonstrate that, distinct from ataxia telangiectasia-like disorder caused by MRE11 pathogenic missense or nonsense variants, mice and cultured cells expressing low MRE11 levels do not display the anticipated defects in ATM activation. Our findings indicate that ATM signaling can be supported by very low levels of the MRN complex and imply that defective ATM activation results from perturbation of MRN function caused by specific hypomorphic disease mutations. These distinct phenotypic outcomes underline the importance of understanding the impact of specific pathogenic MRE11 variants, which may help direct appropriate early surveillance for patients with these complicated disorders in a clinical setting.

3.
Blood ; 142(25): 2159-2174, 2023 12 21.
Article in English | MEDLINE | ID: mdl-37616559

ABSTRACT

ABSTRACT: Activated Notch signaling is highly prevalent in T-cell acute lymphoblastic leukemia (T-ALL), but pan-Notch inhibitors showed excessive toxicity in clinical trials. To find alternative ways to target Notch signals, we investigated cell division cycle 73 (Cdc73), which is a Notch cofactor and key component of the RNA polymerase-associated transcriptional machinery, an emerging target in T-ALL. Although we confirmed previous work that CDC73 interacts with NOTCH1, we also found that the interaction in T-ALL was context-dependent and facilitated by the transcription factor ETS1. Using mouse models, we showed that Cdc73 is important for Notch-induced T-cell development and T-ALL maintenance. Mechanistically, chromatin and nascent gene expression profiling showed that Cdc73 intersects with Ets1 and Notch at chromatin within enhancers to activate expression of known T-ALL oncogenes through its enhancer functions. Cdc73 also intersects with these factors within promoters to activate transcription of genes that are important for DNA repair and oxidative phosphorylation through its gene body functions. Consistently, Cdc73 deletion induced DNA damage and apoptosis and impaired mitochondrial function. The CDC73-induced DNA repair expression program co-opted by NOTCH1 is more highly expressed in T-ALL than in any other cancer. These data suggest that Cdc73 might induce a gene expression program that was eventually intersected and hijacked by oncogenic Notch to augment proliferation and mitigate the genotoxic and metabolic stresses of elevated Notch signaling. Our report supports studying factors such as CDC73 that intersect with Notch to derive a basic scientific understanding on how to combat Notch-dependent cancers without directly targeting the Notch complex.


Subject(s)
5'-Nucleotidase , Leukemia, T-Cell , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Animals , Mice , Cell Line, Tumor , Chromatin , DNA Damage/genetics , Leukemia, T-Cell/genetics , Leukemia, T-Cell/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Transcription Factors/genetics , 5'-Nucleotidase/genetics , 5'-Nucleotidase/metabolism
4.
bioRxiv ; 2023 Feb 04.
Article in English | MEDLINE | ID: mdl-36711472

ABSTRACT

Activated Notch signaling is highly prevalent in T-cell acute lymphoblastic leukemia (T-ALL) but pan-Notch inhibitors were toxic in clinical trials. To find alternative ways to target Notch signals, we investigated Cell division cycle 73 (Cdc73), which is a Notch cofactor and component of transcriptional machinery, a potential target in T-ALL. While we confirmed previous work that CDC73 interacts with NOTCH1, we also found that the interaction in T-ALL was context-dependent and facilitated by the lymphoid transcription factor ETS1. Using mouse models, we showed that Cdc73 is important for Notch-induced T-cell development and T-ALL maintenance. Mechanistically, Cdc73, Ets1, and Notch intersect chromatin at promoters and enhancers to activate oncogenes and genes that are important for DNA repair and oxidative phosphorylation. Consistently, Cdc73 deletion in T-ALL cells induced DNA damage and impaired mitochondrial function. Our data suggests that Cdc73 might promote a gene expression program that was eventually intersected by Notch to mitigate the genotoxic and metabolic stresses of elevated Notch signaling. We also provide mechanistic support for testing inhibitors of DNA repair, oxidative phosphorylation, and transcriptional machinery. Inhibiting pathways like Cdc73 that intersect with Notch at chromatin might constitute a strategy to weaken Notch signals without directly targeting the Notch complex.

5.
Sci Rep ; 11(1): 4268, 2021 02 19.
Article in English | MEDLINE | ID: mdl-33608602

ABSTRACT

Ataxia-telangiectasia (A-T) is an autosomal recessive, multisystem disorder characterized by cerebellar degeneration, cancer predisposition, and immune system defects. A major cause of mortality in A-T patients is severe pulmonary disease; however, the underlying causes of the lung complications are poorly understood, and there are currently no curative therapeutic interventions. In this study, we examined the lung phenotypes caused by ATM-deficient immune cells using a mouse model of A-T pulmonary disease. In response to acute lung injury, ATM-deficiency causes decreased survival, reduced blood oxygen saturation, elevated neutrophil recruitment, exaggerated and prolonged inflammatory responses and excessive lung injury compared to controls. We found that ATM null bone marrow adoptively transferred to WT recipients induces similar phenotypes that culminate in impaired lung function. Moreover, we demonstrated that activated ATM-deficient macrophages exhibit significantly elevated production of harmful reactive oxygen and nitrogen species and pro-inflammatory cytokines. These findings indicate that ATM-deficient immune cells play major roles in causing the lung pathologies in A-T. Based on these results, we examined the impact of inhibiting the aberrant inflammatory responses caused by ATM-deficiency with reparixin, a CXCR1/CXCR2 chemokine receptor antagonist. We demonstrated that reparixin treatment reduces neutrophil recruitment, edema and tissue damage in ATM mutant lungs. Thus, our findings indicate that targeted inhibition of CXCR1/CXCR2 attenuates pulmonary phenotypes caused by ATM-deficiency and suggest that this treatment approach represents a viable therapeutic strategy for A-T lung disease.


Subject(s)
Ataxia Telangiectasia/complications , Ataxia Telangiectasia/genetics , Biomarkers , Disease Susceptibility , Inflammation Mediators/metabolism , Lung Diseases/etiology , Lung Diseases/metabolism , Acute Lung Injury/etiology , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/genetics , Bleomycin/adverse effects , Cytokines/metabolism , DNA Damage , DNA Repair , Disease Models, Animal , Lung Diseases/mortality , Lung Diseases/pathology , Mice , Phenotype , Prognosis
6.
Cancer Res ; 77(19): 5327-5338, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28819025

ABSTRACT

Hypomorphic mutations in the genes encoding the MRE11/RAD50/NBS1 (MRN) DNA repair complex lead to cancer-prone syndromes. MRN binds DNA double-strand breaks, where it functions in repair and triggers cell-cycle checkpoints via activation of the ataxia-telangiectasia mutated kinase. To gain understanding of MRN in cancer, we engineered mice with B lymphocytes lacking MRN, or harboring MRN in which MRE11 lacks nuclease activities. Both forms of MRN deficiency led to hallmarks of cancer, including oncogenic translocations involving c-Myc and the immunoglobulin locus. These preneoplastic B lymphocytes did not progress to detectable B lineage lymphoma, even in the absence of p53. Moreover, Mre11 deficiencies prevented tumorigenesis in a mouse model strongly predisposed to spontaneous B-cell lymphomas. Our findings indicate that MRN cannot be considered a standard tumor suppressor and instead imply that nuclease activities of MRE11 are required for oncogenesis. Inhibition of MRE11 nuclease activity increased DNA damage and selectively induced apoptosis in cells overexpressing oncogenes, suggesting MRE11 serves an important role in countering oncogene-induced replication stress. Thus, MRE11 may offer a target for cancer therapeutic development. More broadly, our work supports the idea that subtle enhancements of endogenous genome instability can exceed the tolerance of cancer cells and be exploited for therapeutic ends. Cancer Res; 77(19); 5327-38. ©2017 AACR.


Subject(s)
B-Lymphocytes/pathology , Cell Transformation, Neoplastic/pathology , DNA Repair Enzymes/physiology , DNA Replication , DNA-Binding Proteins/physiology , Lymphoma, B-Cell/pathology , Proto-Oncogene Proteins c-myc/metabolism , ATP-Binding Cassette Transporters/physiology , Acid Anhydride Hydrolases , Animals , Apoptosis , Ataxia Telangiectasia Mutated Proteins/metabolism , B-Lymphocytes/metabolism , Cell Cycle Proteins/physiology , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , DNA Breaks, Double-Stranded , DNA Repair , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Genomic Instability , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , MRE11 Homologue Protein , Mice , Mutation , Nuclear Proteins/physiology , Oncogenes , Proto-Oncogene Proteins c-myc/genetics
7.
Mol Cell Biol ; 37(12)2017 06 15.
Article in English | MEDLINE | ID: mdl-28320875

ABSTRACT

Allelic exclusion describes the essential immunological process by which feedback repression of sequential DNA rearrangements ensures that only one autosome expresses a functional T or B cell receptor. In wild-type mammals, approximately 60% of cells have recombined the DNA of one T cell receptor ß (TCRß) V-to-DJ-joined allele in a functional configuration, while the second allele has recombined only the DJ sequences; the other 40% of cells have recombined the V to the DJ segments on both alleles, with only one of the two alleles predicting a functional TCRß protein. Here we report that the transgenic overexpression of GATA3 leads predominantly to biallelic TCRß gene (Tcrb) recombination. We also found that wild-type immature thymocytes can be separated into distinct populations based on intracellular GATA3 expression and that GATA3LO cells had almost exclusively recombined only one Tcrb locus (that predicted a functional receptor sequence), while GATA3HI cells had uniformly recombined both Tcrb alleles (one predicting a functional and the other predicting a nonfunctional rearrangement). These data show that GATA3 abundance regulates the recombination propensity at the Tcrb locus and provide new mechanistic insight into the historic immunological conundrum for how Tcrb allelic exclusion is mediated.


Subject(s)
Alleles , GATA3 Transcription Factor/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Animals , GATA3 Transcription Factor/genetics , Gene Expression Regulation , Gene Ontology , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Mutation/genetics , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, RNA , Spleen/metabolism , Thymocytes/metabolism , V(D)J Recombination/genetics
8.
Hum Mol Genet ; 22(24): 4901-13, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23863462

ABSTRACT

SNM1B/Apollo is a DNA nuclease that has important functions in telomere maintenance and repair of DNA interstrand crosslinks (ICLs) within the Fanconi anemia (FA) pathway. SNM1B is required for efficient localization of key repair proteins, such as the FA protein, FANCD2, to sites of ICL damage and functions epistatically to FANCD2 in cellular survival to ICLs and homology-directed repair. The FA pathway is also activated in response to replication fork stalling. Here, we sought to determine the importance of SNM1B in cellular responses to stalled forks in the absence of a blocking lesion, such as ICLs. We found that depletion of SNM1B results in hypersensitivity to aphidicolin, a DNA polymerase inhibitor that causes replication stress. We observed that the SNM1B nuclease is required for efficient localization of the DNA repair proteins, FANCD2 and BRCA1, to subnuclear foci upon aphidicolin treatment, thereby indicating SNM1B facilitates direct repair of stalled forks. Consistent with a role for SNM1B subsequent to recognition of the lesion, we found that SNM1B is dispensable for upstream events, including activation of ATR-dependent signaling and localization of RPA, γH2AX and the MRE11/RAD50/NBS1 complex to aphidicolin-induced foci. We determined that a major consequence of SNM1B depletion is a marked increase in spontaneous and aphidicolin-induced chromosomal gaps and breaks, including breakage at common fragile sites. Thus, this study provides evidence that SNM1B functions in resolving replication stress and preventing accumulation of genomic damage.


Subject(s)
Chromosome Fragile Sites , DNA Repair Enzymes/metabolism , DNA Replication , Genomic Instability , Nuclear Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Aphidicolin/pharmacology , BRCA1 Protein/metabolism , Cell Line , Cell Nucleus/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Chromatin/metabolism , DNA Damage , DNA Repair , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/genetics , Exodeoxyribonucleases , Fanconi Anemia Complementation Group D2 Protein/metabolism , Gene Expression , Histones/metabolism , Humans , Multiprotein Complexes/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Replication Protein A/metabolism , Signal Transduction/drug effects , Ubiquitination
9.
Hum Mol Genet ; 21(19): 4225-36, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22736030

ABSTRACT

Glutamine (Q) expansion diseases are a family of degenerative disorders caused by the lengthening of CAG triplet repeats present in the coding sequences of seemingly unrelated genes whose mutant proteins drive pathogenesis. Despite all the molecular evidence for the genetic basis of these diseases, how mutant poly-Q proteins promote cell death and drive pathogenesis remains controversial. In this report, we show a specific interaction between the mutant androgen receptor (AR), a protein associated with spinal and bulbar muscular atrophy (SBMA), and the nuclear protein PTIP (Pax Transactivation-domain Interacting Protein), a protein with an unusually long Q-rich domain that functions in DNA repair. Upon exposure to ionizing radiation, PTIP localizes to nuclear foci that are sites of DNA damage and repair. However, the expression of poly-Q AR sequesters PTIP away from radiation-induced nuclear foci. This results in sensitivity to DNA-damaging agents and chromosomal instabilities. In a mouse model of SBMA, evidence for DNA damage is detected in muscle cell nuclei and muscular atrophy is accelerated when one copy of the gene encoding PTIP is removed. These data provide a new paradigm for understanding the mechanisms of cellular degeneration observed in poly-Q expansion diseases.


Subject(s)
Bulbo-Spinal Atrophy, X-Linked/genetics , Bulbo-Spinal Atrophy, X-Linked/metabolism , Carrier Proteins/metabolism , DNA Repair , Genomic Instability , Nuclear Proteins/metabolism , Peptides/genetics , Receptors, Androgen/metabolism , Trinucleotide Repeat Expansion , Animals , Carrier Proteins/genetics , DNA-Binding Proteins , Humans , Mice , Mice, Knockout , Nuclear Proteins/genetics , Peptides/metabolism , Protein Binding , Protein Structure, Tertiary , Receptors, Androgen/genetics
10.
Hum Mol Genet ; 20(13): 2549-59, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21478198

ABSTRACT

Fanconi anemia (FA) is an inherited chromosomal instability disorder characterized by childhood aplastic anemia, developmental abnormalities and cancer predisposition. One of the hallmark phenotypes of FA is cellular hypersensitivity to agents that induce DNA interstrand crosslinks (ICLs), such as mitomycin C (MMC). FA is caused by mutation in at least 14 genes which function in the resolution of ICLs during replication. The FA proteins act within the context of a protein network in coordination with multiple repair factors that function in distinct pathways. SNM1B/Apollo is a member of metallo-ß-lactamase/ßCASP family of nucleases and has been demonstrated to function in ICL repair. However, the relationship between SNM1B and the FA protein network is not known. In the current study, we establish that SNM1B functions epistatically to the central FA factor, FANCD2, in cellular survival after ICL damage and homology-directed repair of DNA double-strand breaks. We also demonstrate that MMC-induced chromosomal anomalies are increased in SNM1B-depleted cells, and this phenotype is not further exacerbated upon depletion of either FANCD2 or another key FA protein, FANCI. Furthermore, we find that SNM1B is required for proper localization of critical repair factors, including FANCD2, BRCA1 and RAD51, to MMC-induced subnuclear foci. Our findings demonstrate that SNM1B functions within the FA pathway during the repair of ICL damage.


Subject(s)
DNA Damage , DNA Repair Enzymes/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Fanconi Anemia/enzymology , Nuclear Proteins/metabolism , Signal Transduction , Alkylating Agents/pharmacology , Chromosomal Instability/drug effects , DNA Breaks, Double-Stranded , DNA Damage/drug effects , DNA Repair/drug effects , DNA Repair Enzymes/genetics , Exodeoxyribonucleases , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group Proteins/genetics , Gene Expression Regulation , Gene Knockdown Techniques , HeLa Cells , Humans , Mitomycin/pharmacology , Nuclear Proteins/genetics , Protein Binding/drug effects , Signal Transduction/genetics
11.
Hum Mol Genet ; 20(4): 806-19, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21147755

ABSTRACT

The Artemis gene encodes a DNA nuclease that plays important roles in non-homologous end-joining (NHEJ), a major double-strand break (DSB) repair pathway in mammalian cells. NHEJ factors repair general DSBs as well as programmed breaks generated during the lymphoid-specific DNA rearrangement, V(D)J recombination, which is required for lymphocyte development. Mutations that inactivate Artemis cause a human severe combined immunodeficiency syndrome associated with cellular radiosensitivity. In contrast, hypomorphic Artemis mutations result in combined immunodeficiency syndromes of varying severity, but, in addition, are hypothesized to predispose to lymphoid malignancy. To elucidate the distinct molecular defects caused by hypomorphic compared with inactivating Artemis mutations, we examined tumor predisposition in a mouse model harboring a targeted partial loss-of-function disease allele. We find that, in contrast to Artemis nullizygosity, the hypomorphic mutation leads to increased aberrant intra- and interchromosomal V(D)J joining events. We also observe that dysfunctional Artemis activity combined with p53 inactivation predominantly predisposes to thymic lymphomas harboring clonal translocations distinct from those observed in Artemis nullizygosity. Thus, the Artemis hypomorphic allele results in unique molecular defects, tumor spectrum and oncogenic chromosomal rearrangements. Our findings have significant implications for disease outcomes and treatment of patients with different Artemis mutations.


Subject(s)
Alleles , Chromosome Aberrations , Gene Rearrangement , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/metabolism , Animals , DNA Damage , DNA-Binding Proteins , Disease Models, Animal , Endonucleases , Humans , Lymphoma/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Spectral Karyotyping , Survival Analysis , Tumor Suppressor Protein p53/genetics
12.
J Exp Med ; 207(7): 1541-54, 2010 Jul 05.
Article in English | MEDLINE | ID: mdl-20547827

ABSTRACT

The contribution of B cells to the pathology of Omenn syndrome and leaky severe combined immunodeficiency (SCID) has not been previously investigated. We have studied a mut/mut mouse model of leaky SCID with a homozygous Rag1 S723C mutation that impairs, but does not abrogate, V(D)J recombination activity. In spite of a severe block at the pro-B cell stage and profound B cell lymphopenia, significant serum levels of immunoglobulin (Ig) G, IgM, IgA, and IgE and a high proportion of Ig-secreting cells were detected in mut/mut mice. Antibody responses to trinitrophenyl (TNP)-Ficoll and production of high-affinity antibodies to TNP-keyhole limpet hemocyanin were severely impaired, even after adoptive transfer of wild-type CD4(+) T cells. Mut/mut mice produced high amounts of low-affinity self-reactive antibodies and showed significant lymphocytic infiltrates in peripheral tissues. Autoantibody production was associated with impaired receptor editing and increased serum B cell-activating factor (BAFF) concentrations. Autoantibodies and elevated BAFF levels were also identified in patients with Omenn syndrome and leaky SCID as a result of hypomorphic RAG mutations. These data indicate that the stochastic generation of an autoreactive B cell repertoire, which is associated with defects in central and peripheral checkpoints of B cell tolerance, is an important, previously unrecognized, aspect of immunodeficiencies associated with hypomorphic RAG mutations.


Subject(s)
Antibody-Producing Cells/immunology , Antibody-Producing Cells/pathology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Homeodomain Proteins/immunology , Immune Tolerance/immunology , Animals , Antibody Formation/immunology , Autoantibodies/blood , B-Cell Activating Factor/blood , Cell Proliferation , Homeodomain Proteins/genetics , Humans , Immunity/immunology , Immunization , Immunologic Deficiency Syndromes/blood , Immunologic Deficiency Syndromes/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation/genetics , Spleen/immunology , Spleen/pathology
13.
Nat Struct Mol Biol ; 16(8): 808-13, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19633670

ABSTRACT

The Mre11-Rad50-NBS1 (MRN) complex has many roles in response to DNA double-strand breaks, but its functions in repair by nonhomologous end joining (NHEJ) pathways are poorly understood. We have investigated requirements for MRN in class switch recombination (CSR), a programmed DNA rearrangement in B lymphocytes that requires NHEJ. To this end, we have engineered mice that lack the entire MRN complex in B lymphocytes or that possess an intact complex that harbors mutant Mre11 lacking DNA nuclease activities. MRN deficiency confers a strong defect in CSR, affecting both the classic and the alternative NHEJ pathways. In contrast, absence of Mre11 nuclease activities causes a milder phenotype, revealing a separation of function within the complex. We propose a model in which MRN stabilizes distant breaks and processes DNA termini to facilitate repair by both the classical and alternative NHEJ pathways.


Subject(s)
B-Lymphocytes/metabolism , DNA Repair , Immunoglobulin Class Switching , Signal Transduction/physiology , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Acid Anhydride Hydrolases , Adaptor Proteins, Signal Transducing , Animals , Ataxia Telangiectasia Mutated Proteins , B-Lymphocytes/cytology , Base Sequence , Blotting, Western , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Cells, Cultured , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Flow Cytometry , Histones/genetics , Histones/metabolism , Immunoglobulin Heavy Chains/genetics , In Situ Hybridization, Fluorescence , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , MRE11 Homologue Protein , Male , Mice , Mice, Knockout , Molecular Sequence Data , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Recombination, Genetic , Signal Transduction/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
14.
J Exp Med ; 206(4): 893-908, 2009 Apr 13.
Article in English | MEDLINE | ID: mdl-19349461

ABSTRACT

Artemis was initially discovered as the gene inactivated in human radiosensitive T(-)B(-) severe combined immunodeficiency, a syndrome characterized by the absence of B and T lymphocytes and cellular hypersensitivity to ionizing radiation. Hypomorphic Artemis alleles have also been identified in patients and are associated with combined immunodeficiencies of varying severity. We examine the molecular mechanisms underlying a syndrome of partial immunodeficiency caused by a hypomorphic Artemis allele using the mouse as a model system. This mutation, P70, leads to premature translation termination that deletes a large portion of a nonconserved C terminus. We find that homozygous Artemis-P70 mice exhibit reduced numbers of B and T lymphocytes, thereby recapitulating the patient phenotypes. The hypomorphic mutation results in impaired end processing during the lymphoid-specific DNA rearrangement known as V(D)J recombination, defective double-strand break repair, and increased chromosomal instability. Biochemical analyses reveal that the Artemis-P70 mutant protein interacts with the DNA-dependent protein kinase catalytic subunit and retains significant, albeit reduced, exo- and endonuclease activities but does not undergo phosphorylation. Together, our findings indicate that the Artemis C terminus has critical in vivo functions in ensuring efficient V(D)J rearrangements and maintaining genome integrity.


Subject(s)
Genome, Human , Genome , Severe Combined Immunodeficiency/genetics , Animals , DNA Damage , Disease Models, Animal , Gene Rearrangement/immunology , Humans , Mice , Mice, Transgenic , Mutation , RNA/genetics , RNA/isolation & purification , T-Lymphocytes/immunology
15.
Mol Cell Biol ; 29(11): 3163-72, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19332554

ABSTRACT

Nonhomologous end-joining represents the major pathway used by human cells to repair DNA double-strand breaks. It relies on the XRCC4/DNA ligase IV complex to reseal DNA strands. Here we report the high-resolution crystal structure of human XRCC4 bound to the carboxy-terminal tandem BRCT repeat of DNA ligase IV. The structure differs from the homologous Saccharomyces cerevisiae complex and reveals an extensive DNA ligase IV binding interface formed by a helix-loop-helix structure within the inter-BRCT linker region, as well as significant interactions involving the second BRCT domain, which induces a kink in the tail region of XRCC4. We further demonstrate that interaction with the second BRCT domain of DNA ligase IV is necessary for stable binding to XRCC4 in cells, as well as to achieve efficient dominant-negative effects resulting in radiosensitization after ectopic overexpression of DNA ligase IV fragments in human fibroblasts. Together our findings provide unanticipated insight for understanding the physical and functional architecture of the nonhomologous end-joining ligation complex.


Subject(s)
DNA Ligases/chemistry , DNA Ligases/metabolism , DNA Repair , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Amino Acid Sequence , Binding, Competitive , Cell Line , DNA Breaks, Double-Stranded , DNA Ligase ATP , DNA Repair Enzymes/metabolism , Down-Regulation , Humans , Molecular Sequence Data , Protein Binding , Protein Stability , Protein Structure, Secondary , Protein Structure, Tertiary , Radiation Tolerance , Recombination, Genetic/genetics , Structural Homology, Protein , Structure-Activity Relationship
16.
Blood ; 113(13): 2965-75, 2009 Mar 26.
Article in English | MEDLINE | ID: mdl-19126872

ABSTRACT

The RAG1/2 endonuclease initiates programmed DNA rearrangements in progenitor lymphocytes by generating double-strand breaks at specific recombination signal sequences. This process, known as V(D)J recombination, assembles the vastly diverse antigen receptor genes from numerous V, D, and J coding segments. In vitro biochemical and cellular transfection studies suggest that RAG1/2 may also play postcleavage roles by forming complexes with the recombining ends to facilitate DNA end processing and ligation. In the current study, we examine the in vivo consequences of a mutant form of RAG1, RAG1-S723C, that is proficient for DNA cleavage, yet exhibits defects in postcleavage complex formation and end joining in vitro. We generated a knockin mouse model harboring the RAG1-S723C hypomorphic mutation and examined the immune system in this fully in vivo setting. RAG1-S723C homozygous mice exhibit impaired lymphocyte development and decreased V(D)J rearrangements. Distinct from RAG nullizygosity, the RAG1-S723C hypomorph results in aberrant DNA double-strand breaks within rearranging loci. RAG1-S723C also predisposes to thymic lymphomas associated with chromosomal translocations in a p53 mutant background, and heterozygosity for the mutant allele accelerates age-associated immune system dysfunction. Thus, our study provides in vivo evidence that implicates aberrant RAG1/2 activity in lymphoid tumor development and premature immunosenescence.


Subject(s)
Gene Rearrangement/genetics , Homeodomain Proteins/genetics , Mutation, Missense , Severe Combined Immunodeficiency/genetics , Aging/genetics , Aging/immunology , Amino Acid Substitution/physiology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Gene Knock-In Techniques , Homozygote , Lymphoma/genetics , Lymphoma/immunology , Mice , Mice, Transgenic , Mutation, Missense/physiology , Phenotype , Severe Combined Immunodeficiency/immunology , Severe Combined Immunodeficiency/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Thymus Neoplasms/genetics , Thymus Neoplasms/immunology , VDJ Exons
17.
Cell ; 135(1): 85-96, 2008 Oct 03.
Article in English | MEDLINE | ID: mdl-18854157

ABSTRACT

The Mre11/Rad50/NBS1 (MRN) complex maintains genomic stability by bridging DNA ends and initiating DNA damage signaling through activation of the ATM kinase. Mre11 possesses DNA nuclease activities that are highly conserved in evolution but play unknown roles in mammals. To define the functions of Mre11, we engineered targeted mouse alleles that either abrogate nuclease activities or inactivate the entire MRN complex. Mre11 nuclease deficiency causes a striking array of phenotypes indistinguishable from the absence of MRN, including early embryonic lethality and dramatic genomic instability. We identify a crucial role for the nuclease activities in homology-directed double-strand-break repair and a contributing role in activating the ATR kinase. However, the nuclease activities are not required to activate ATM after DNA damage or telomere deprotection. Therefore, nucleolytic processing by Mre11 is an essential function of fundamental importance in DNA repair, distinct from MRN control of ATM signaling.


Subject(s)
DNA Repair Enzymes/metabolism , DNA Repair , DNA-Binding Proteins/metabolism , Genomic Instability , Amino Acid Sequence , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Line, Transformed , Cell Proliferation , DNA Breaks, Double-Stranded , DNA Damage , DNA Repair Enzymes/chemistry , DNA-Binding Proteins/chemistry , Fibroblasts/metabolism , MRE11 Homologue Protein , Mice , Protein Serine-Threonine Kinases/metabolism , Recombination, Genetic , Telomere/metabolism , Tumor Suppressor Proteins/metabolism
18.
Proc Natl Acad Sci U S A ; 105(27): 9302-6, 2008 Jul 08.
Article in English | MEDLINE | ID: mdl-18599436

ABSTRACT

Upon DNA damage, histone H2AX is phosphorylated by ataxia-telangiectasia mutated (ATM) and other phosphoinositide 3-kinase-related protein kinases. To elucidate further the potential overlapping and unique functions of ATM and H2AX, we asked whether they have synergistic functions in the development and maintenance of genomic stability by inactivating both genes in mouse germ line. Combined ATM/H2AX deficiency caused embryonic lethality and dramatic cellular genomic instability. Mechanistically, severe genomic instability in the double-deficient cells is associated with a requirement for H2AX to repair oxidative DNA damage resulting from ATM deficiency. We discuss these findings in the context of synergies between ATM and other repair factors.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Genomic Instability , Histones/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Proliferation , DNA Damage , DNA Repair , Embryo Loss/metabolism , Embryo, Mammalian/abnormalities , Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Mice , Pregnancy , Reactive Oxygen Species/metabolism
19.
Nature ; 446(7132): 208-12, 2007 Mar 08.
Article in English | MEDLINE | ID: mdl-17344853

ABSTRACT

Long interspersed element-1 (LINE-1 or L1) elements are abundant, non-long-terminal-repeat (non-LTR) retrotransposons that comprise approximately 17% of human DNA. The average human genome contains approximately 80-100 retrotransposition-competent L1s (ref. 2), and they mobilize by a process that uses both the L1 endonuclease and reverse transcriptase, termed target-site primed reverse transcription. We have previously reported an efficient, endonuclease-independent L1 retrotransposition pathway (EN(i)) in certain Chinese hamster ovary (CHO) cell lines that are defective in the non-homologous end-joining (NHEJ) pathway of DNA double-strand-break repair. Here we have characterized EN(i) retrotransposition events generated in V3 CHO cells, which are deficient in DNA-dependent protein kinase catalytic subunit (DNA-PKcs) activity and have both dysfunctional telomeres and an NHEJ defect. Notably, approximately 30% of EN(i) retrotransposition events insert in an orientation-specific manner adjacent to a perfect telomere repeat (5'-TTAGGG-3'). Similar insertions were not detected among EN(i) retrotransposition events generated in controls or in XR-1 CHO cells deficient for XRCC4, an NHEJ factor that is required for DNA ligation but has no known function in telomere maintenance. Furthermore, transient expression of a dominant-negative allele of human TRF2 (also called TERF2) in XRCC4-deficient XR-1 cells, which disrupts telomere capping, enables telomere-associated EN(i) retrotransposition events. These data indicate that L1s containing a disabled endonuclease can use dysfunctional telomeres as an integration substrate. The findings highlight similarities between the mechanism of EN(i) retrotransposition and the action of telomerase, because both processes can use a 3' OH for priming reverse transcription at either internal DNA lesions or chromosome ends. Thus, we propose that EN(i) retrotransposition is an ancestral mechanism of RNA-mediated DNA repair associated with non-LTR retrotransposons that may have been used before the acquisition of an endonuclease domain.


Subject(s)
Long Interspersed Nucleotide Elements/genetics , Mutagenesis, Insertional/genetics , Retroelements/genetics , Telomere/genetics , Animals , Base Sequence , Cell Line , Chromosomal Instability/genetics , Cricetinae , Cricetulus , Endonucleases/deficiency , Endonucleases/genetics , Endonucleases/metabolism , Humans , Polymerase Chain Reaction/methods
20.
Cell ; 124(2): 260-2, 2006 Jan 27.
Article in English | MEDLINE | ID: mdl-16439201

ABSTRACT

A major pathway for repair of DNA double-strand breaks is nonhomologous end-joining (NHEJ). In this issue of Cell, and report the discovery of a new NHEJ factor called Cernunnos-XLF. Both groups report that this protein is mutated in a rare inherited human syndrome characterized by severe immunodeficiency, developmental delay, and hypersensitivity to agents that cause DNA double-strand breaks.


Subject(s)
DNA Damage , DNA Repair , DNA-Binding Proteins/physiology , Animals , DNA Repair Enzymes , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...