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










Publication year range
1.
DNA Repair (Amst) ; 68: 34-40, 2018 08.
Article in English | MEDLINE | ID: mdl-29929045

ABSTRACT

Classical non-homologous end-joining (cNHEJ) is the main pathway for the repair of DNA double strand breaks (DSBs) in mammalian cells. In the absence of c-NHEJ, an alternative end-joining (A-EJ) mechanism resolves DSBs. To date, no A-EJ specific factor has been identified. Instead, this mechanism appears to co-opt proteins involved in more than one DNA repair pathway. These include components of base-excision repair (PARP1/XRCC1/LIG3), interstrand cross-link repair (BRCA1/FANCD2), and DSB response/DNA end-resection (MRE11A/RAD50/RBBP8). To clarify the contribution of these factors to A-EJ, here we examined their expression and recruitment to DSBs in correlation with surrogates of cNHEJ (53BP1) and homologous recombination (RAD51) in cells deficient for the cNHEJ end-ligation component XRCC4. This revealed XRCC4-deficient cells exhibited marked increases in the stability of A-EJ transcripts that result in correspondingly elevated levels of associated proteins, in comparison to WT cells. RAD51 was also increased while 53BP1 was unaffected. Treatment with radiomimetic DSB-inducing drug doxorubicin did not influence these activities. However, FANCD2, BRCA1 and XRCC1 foci, prominently associated with 53BP1 foci and hence DSBs resolved by cNHEJ, were only detected in doxorubicin-treated XRCC4-deficient cells. Strikingly, treatment of XRCC4-deficient cells with the PARP-specific inhibitor Niraparib enhanced A-EJ, and substantially induced 53BP1 transcripts and the numbers of A-EJ-associated 53BP1 DNA damage foci. RAD51 was severely inhibited, and upstream cNHEJ (KU70/KU80/DNA-PKCs/ARTEMIS) transcripts were substantially induced. These latter results were recapitulated in BRCA1-deficient cells, which contrastingly did not affect 53BP1 or PARP1 status irrespective of doxorubicin or Niraparib treatment. Hence A-EJ is regulated transcriptionally, reduced by a higher turnover rate in cNHEJ-proficient cells and sustained but fine-tuned by PARP1 in XRCC4-deficient cells to promote DNA repair and survival. Upstream cNHEJ components are similarly transcriptionally down-modulated by PARP1 and BRCA1 in a manner inversely correlated with HR and mechanistically distinct from A-EJ respectively in cNHEJ-deficient and cNHEJ-proficient settings.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins/genetics , Signal Transduction , Animals , BRCA1 Protein/metabolism , Cells, Cultured , DNA/metabolism , DNA End-Joining Repair/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Gene Knockout Techniques , Humans , Mice , Poly (ADP-Ribose) Polymerase-1/metabolism , Rad51 Recombinase/genetics , Tumor Suppressor p53-Binding Protein 1/genetics
3.
Nat Commun ; 8: 14013, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28094268

ABSTRACT

DNA repair gene defects are found in virtually all human glioblastomas, but the genetic evidence for a direct role remains lacking. Here we demonstrate that combined inactivation of the XRCC4 non-homologous end-joining (NHEJ) DNA repair gene and p53 efficiently induces brain tumours with hallmark characteristics of human proneural/classical glioblastoma. The murine tumours exhibit PTEN loss of function instigated by reduced PTEN mRNA, and increased phosphorylated inactivation and stability as a consequence of aberrantly elevated CK2 provoked by p53 ablation and irrevocably deregulated by NHEJ inactivation. This results in DNA damage-resistant cytoplasmic PTEN and CK2 expression, and the attenuation of DNA repair genes. CK2 inhibition restores PTEN nuclear distribution and DNA repair activities and impairs tumour but not normal cell survival. These observations demonstrate that NHEJ contributes to p53-mediated glioblastoma suppression, and reveal a crucial role for PTEN in the early DNA damage signalling cascade, the inhibition of which promotes tumorigenicity and drug-resistant survival.


Subject(s)
Brain Neoplasms/genetics , DNA End-Joining Repair , Glioblastoma/genetics , PTEN Phosphohydrolase/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cell Line, Tumor , DNA Damage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm , Female , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Male , Mice , PTEN Phosphohydrolase/genetics , Signal Transduction , Tumor Suppressor Protein p53/genetics
4.
J Immunol ; 196(1): 244-55, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26608917

ABSTRACT

Hypomorphic mutations in the nonhomologous end-joining (NHEJ) DNA repair protein DNA ligase IV (LIG4) lead to immunodeficiency with varying severity. In this study, using a murine knock-in model, we investigated the mechanisms underlying abnormalities in class switch recombination (CSR) associated with the human homozygous Lig4 R278H mutation. Previously, we found that despite the near absence of Lig4 end-ligation activity and severely reduced mature B cell numbers, Lig4(R278H/R278H) (Lig4(R/R)) mice exhibit only a partial CSR block, producing near normal IgG1 and IgE but substantially reduced IgG3, IgG2b, and IgA serum levels. In this study, to address the cause of these abnormalities, we assayed CSR in Lig4(R/R) B cells generated via preassembled IgH and IgK V region exons (HL). This revealed that Lig4(R278H) protein levels while intact exhibited a higher turnover rate during activation of switching to IgG3 and IgG2b, as well as delays in CSR kinetics associated with defective proliferation during activation of switching to IgG1 and IgE. Activated Lig4(R/R)HL B cells consistently accumulated high frequencies of activation-induced cytidine deaminase-dependent IgH locus chromosomal breaks and translocations and were more prone to apoptosis, effects that appeared to be p53-independent, as p53 deficiency did not markedly influence these events. Importantly, NHEJ instead of alternative end-joining (A-EJ) was revealed as the predominant mechanism catalyzing robust CSR. Defective CSR was linked to failed NHEJ and residual A-EJ access to unrepaired double-strand breaks. These data firmly demonstrate that Lig4(R278H) activity renders NHEJ to be more error-prone, and they predict increased error-prone NHEJ activity and A-EJ suppression as the cause of the defective B lymphopoiesis in Lig4 patients.


Subject(s)
B-Lymphocytes/immunology , DNA End-Joining Repair/genetics , DNA Ligases/genetics , Eczema/genetics , Growth Disorders/genetics , Intellectual Disability/genetics , Lymphopoiesis/genetics , Microcephaly/genetics , Severe Combined Immunodeficiency/genetics , Animals , Apoptosis/genetics , Apoptosis/immunology , Base Sequence , Cell Proliferation , Cells, Cultured , Cytidine Deaminase/metabolism , DNA Breaks, Double-Stranded , DNA Ligase ATP , Disease Models, Animal , Facies , Gene Knock-In Techniques , Humans , Immunoglobulin A/blood , Immunoglobulin A/genetics , Immunoglobulin A/immunology , Immunoglobulin Class Switching/genetics , Immunoglobulin Class Switching/immunology , Immunoglobulin E/blood , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulins/genetics , Mice , Mice, Knockout , Molecular Sequence Data , Tumor Suppressor Protein p53/genetics
5.
Blood ; 125(5): 852-5, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25477498

ABSTRACT

Mice with T-cell-specific loss of the tumor suppressor gene PTEN early in T-cell ontogeny develop thymic lymphomas that invariably harbor a reciprocal translocation involving the T-cell receptor α/δ locus and c-myc, t(14;15). In addition to its known function as a lipid phosphatase opposing PI3K signaling, PTEN has also been described as playing a prominent role in promoting genomic stability. As a result, it has been uncertain which one(s) of these 2 separable features were required to block the development of lymphoma. Here, using a conditional model in which T cells selectively express 1 phosphatase-dead PTEN mutant (C124S) and maintain 1 null allele, we show that PTEN phosphatase activity is required for preventing the emergence of a malignant T-cell population harboring t(14;15), thus constituting a critical function of PTEN in preventing lymphomagenesis.


Subject(s)
Gene Expression Regulation, Neoplastic , Lymphoma, T-Cell/genetics , PTEN Phosphohydrolase/genetics , T-Lymphocytes/enzymology , Thymus Neoplasms/genetics , Animals , Bone Marrow/enzymology , Bone Marrow/pathology , Chimera/genetics , Chimera/metabolism , Chromosomes, Mammalian , Lymphoma, T-Cell/enzymology , Lymphoma, T-Cell/pathology , Mice , Mice, Knockout , PTEN Phosphohydrolase/deficiency , Protein Isoforms/deficiency , Protein Isoforms/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Signal Transduction , T-Lymphocytes/pathology , Thymus Gland/enzymology , Thymus Gland/pathology , Thymus Neoplasms/enzymology , Thymus Neoplasms/pathology , Translocation, Genetic
6.
Haematologica ; 98(5): 739-43, 2013 May.
Article in English | MEDLINE | ID: mdl-23065515

ABSTRACT

The SET domain is found in histone methyltransferases and other lysine methyltransferases. SET domain-containing proteins such as MLL1 play a critical role in leukemogenesis, while others such as SETD2 may function as a tumor suppressor in breast cancer and renal cell carcinoma. We recently discovered that SETD3, a well-conserved SET domain-containing protein, was involved in a translocation to the immunoglobulin lambda light chain locus in one of the non-homologous end-joining/p53-deficient peripheral B-cell lymphomas. We showed that a truncated mRNA lacking the SET domain sequences in Setd3 gene was highly expressed in the lymphoma. Furthermore, we found that the truncated SET-less protein displayed oncogenic potential while the full length SETD3 protein did not. Finally, SETD3 exhibits histone methyltransferases activity on nucleosomal histone 3 in a SET-domain dependent manner. We propose that this newly identified Setd3 gene may play an important role in carcinogenesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Histone-Lysine N-Methyltransferase/genetics , Animals , Cell Line , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cloning, Molecular , DNA End-Joining Repair , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Methylation , Mice , Tumor Suppressor Protein p53/deficiency
7.
J Immunol ; 186(4): 2336-43, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21242524

ABSTRACT

In the absence of core nonhomologous end-joining (NHEJ) factors, Ab gene class-switch recombination (CSR) uses an alternative end-joining (A-EJ) pathway to recombine switch (S) region DNA breaks. Previous reports showing decreased S-junction microhomologies in MSH2-deficient mice and an exonuclease 1 (EXO1) role in yeast microhomology-mediated end joining suggest that mismatch repair (MMR) proteins might influence A-EJ-mediated CSR. We have directly investigated whether MMR proteins collectively or differentially influence the A-EJ mechanism of CSR by analyzing CSR in mice deficient in both XRCC4 and individual MMR proteins. We find CSR is reduced and that Igh locus chromosome breaks are reduced in the MMR/XRCC4 double-deficient B cells compared with B cells deficient in XRCC4 alone, suggesting MMR proteins function upstream of double-strand break formation to influence CSR efficiency in these cells. Our results show that MLH1, EXO1, and MSH2 are all important for efficient A-EJ-mediated CSR, and we propose that MMR proteins convert DNA nicks and point mutations into dsDNA breaks for both C-NHEJ and A-EJ pathways of CSR. We also find Mlh1-XRCC4(-) B cells have an increased frequency of direct S junctions, suggesting that MLH1 proteins may have additional functions that influence A-EJ-mediated CSR.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , B-Lymphocyte Subsets/metabolism , DNA Repair/genetics , DNA-Binding Proteins/deficiency , Exodeoxyribonucleases/physiology , Immunoglobulin Class Switching/genetics , MutS Homolog 2 Protein/physiology , Nuclear Proteins/physiology , Animals , Cells, Cultured , DNA Breaks, Double-Stranded , DNA Damage , DNA-Binding Proteins/genetics , Deoxyribonucleases, Type II Site-Specific , Mice , Mice, Knockout , Mice, Transgenic , MutL Protein Homolog 1 , Point Mutation
8.
J Exp Med ; 207(2): 417-27, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20142431

ABSTRACT

The classical nonhomologous end-joining (C-NHEJ) DNA double-strand break (DSB) repair pathway employs the Ku70/80 complex (Ku) for DSB recognition and the XRCC4/DNA ligase 4 (Lig4) complex for ligation. During IgH class switch recombination (CSR) in B lymphocytes, switch (S) region DSBs are joined by C-NHEJ to form junctions either with short microhomologies (MHs; "MH-mediated" joins) or no homologies ("direct" joins). In the absence of XRCC4 or Lig4, substantial CSR occurs via "alternative" end-joining (A-EJ) that generates largely MH-mediated joins. Because upstream C-NHEJ components remain in XRCC4- or Lig4-deficient B cells, residual CSR might be catalyzed by C-NHEJ using a different ligase. To address this, we have assayed for CSR in B cells deficient for Ku70, Ku80, or both Ku70 and Lig4. Ku70- or Ku80-deficient B cells have reduced, but still substantial, CSR. Strikingly, B cells deficient for both Ku plus Lig4 undergo CSR similarly to Ku-deficient B cells, firmly demonstrating that an A-EJ pathway distinct from C-NHEJ can catalyze CSR end-joining. Ku-deficient or Ku- plus Lig4-deficient B cells are also biased toward MH-mediated CSR joins; but, in contrast to XRCC4- or Lig4-deficient B cells, generate substantial numbers of direct CSR joins. Our findings suggest that more than one form of A-EJ can function in CSR.


Subject(s)
Antigens, Nuclear/metabolism , B-Lymphocytes/metabolism , DNA Breaks, Double-Stranded , DNA Ligases/metabolism , DNA Repair , DNA-Binding Proteins/metabolism , Immunoglobulin Class Switching , Immunoglobulins/genetics , Animals , Antigens, Nuclear/immunology , B-Lymphocytes/immunology , DNA Ligase ATP , DNA Ligases/immunology , DNA-Binding Proteins/immunology , Immunoglobulins/metabolism , Ku Autoantigen , Mice
9.
Proc Natl Acad Sci U S A ; 107(7): 3024-9, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20133615

ABSTRACT

DNA ligase IV (LIG4) is an essential component of the nonhomologous end-joining (NHEJ) repair pathway and plays a key role in V(D)J recombination. Hypomorphic LIG4 mutations in humans are associated with increased cellular radiosensitivity, microcephaly, facial dysmorphisms, growth retardation, developmental delay, and a variable degree of immunodeficiency. We have generated a knock-in mouse model with a homozygous Lig4 R278H mutation that corresponds to the first LIG4 mutation reported in humans. The phenotype of homozygous mutant mice Lig4(R278H/R278H) (Lig4(R/R)) includes growth retardation, a decreased life span, a severe cellular sensitivity to ionizing radiation, and a very severe, but incomplete block in T and B cell development. Peripheral T lymphocytes show an activated and anergic phenotype, reduced viability, and a restricted repertoire, reminiscent of human leaky SCID. Genomic instability is associated with a high rate of thymic tumor development. Finally, Lig4(R/R) mice spontaneously produce low-affinity antibodies that include autoreactive specificities, but are unable to mount high-affinity antibody responses. These findings highlight the importance of LIG4 in lymphocyte development and function, and in genomic stability maintenance, and provide a model for the complex phenotype of LIG4 syndrome in humans.


Subject(s)
Abnormalities, Multiple/genetics , Antibody Formation/genetics , DNA Ligases/genetics , Developmental Disabilities/genetics , Disease Models, Animal , Mutation, Missense/genetics , Severe Combined Immunodeficiency/genetics , Animals , Apoptosis/immunology , Blotting, Southern , Child , DNA Ligase ATP , DNA Ligases/immunology , Flow Cytometry , Humans , Immunoglobulins/blood , Immunophenotyping , Mice , Mutation, Missense/immunology , Syndrome
10.
Proc Natl Acad Sci U S A ; 107(7): 3034-9, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20133803

ABSTRACT

Class switch recombination (CSR) in B lymphocytes is initiated by introduction of multiple DNA double-strand breaks (DSBs) into switch (S) regions that flank immunoglobulin heavy chain (IgH) constant region exons. CSR is completed by joining a DSB in the donor S mu to a DSB in a downstream acceptor S region (e.g., S gamma1) by end-joining. In normal cells, many CSR junctions are mediated by classical nonhomologous end-joining (C-NHEJ), which employs the Ku70/80 complex for DSB recognition and XRCC4/DNA ligase 4 for ligation. Alternative end-joining (A-EJ) mediates CSR, at reduced levels, in the absence of C-NHEJ, even in combined absence of Ku70 and ligase 4, demonstrating an A-EJ pathway totally distinct from C-NHEJ. Multiple DSBs are introduced into S mu during CSR, with some being rejoined or joined to each other to generate internal switch deletions (ISDs). In addition, S-region DSBs can be joined to other chromosomes to generate translocations, the level of which is increased by absence of a single C-NHEJ component (e.g., XRCC4). We asked whether ISD and S-region translocations occur in the complete absence of C-NHEJ (e.g., in Ku70/ligase 4 double-deficient B cells). We found, unexpectedly, that B-cell activation for CSR generates substantial ISD in both S mu and S gamma1 and that ISD in both is greatly increased by the absence of C-NHEJ. IgH chromosomal translocations to the c-myc oncogene also are augmented in the combined absence of Ku70 and ligase 4. We discuss the implications of these findings for A-EJ in normal and abnormal DSB repair.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair/immunology , Immunoglobulin Class Switching/genetics , Immunoglobulin Switch Region/genetics , Translocation, Genetic/genetics , Animals , Antigens, Nuclear/genetics , B-Lymphocytes/immunology , Base Sequence , Blotting, Southern , DNA Ligase ATP , DNA Ligases/genetics , DNA Primers/genetics , DNA-Binding Proteins/genetics , Genes, myc/genetics , Immunoglobulin Class Switching/immunology , Immunoglobulin Heavy Chains/genetics , In Situ Hybridization, Fluorescence , Ku Autoantigen , Mice , Mice, Knockout , Molecular Sequence Data , Translocation, Genetic/immunology
11.
Nature ; 462(7274): 803-7, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-20010689

ABSTRACT

B-cell malignancies, such as human Burkitt's lymphoma, often contain translocations that link c-myc or other proto-oncogenes to the immunoglobulin heavy chain locus (IgH, encoded by Igh). The nature of elements that activate oncogenes within such translocations has been a long-standing question. Translocations within Igh involve DNA double-strand breaks initiated either by the RAG1/2 endonuclease during variable, diversity and joining gene segment (V(D)J) recombination, or by activation-induced cytidine deaminase (AID, also known as AICDA) during class switch recombination (CSR). V(D)J recombination in progenitor B (pro-B) cells assembles Igh variable region exons upstream of mu constant region (Cmu) exons, which are the first of several sets of C(H) exons ('C(H) genes') within a C(H) locus that span several hundred kilobases (kb). In mature B cells, CSR deletes Cmu and replaces it with a downstream C(H) gene. An intronic enhancer (iEmu) between the variable region exons and Cmu promotes V(D)J recombination in developing B cells. Furthermore, the Igh 3' regulatory region (Igh3'RR) lies downstream of the C(H) locus and modulates CSR by long-range transcriptional enhancement of C(H) genes. Transgenic mice bearing iEmu or Igh3'RR sequences fused to c-myc are predisposed to B lymphomas, demonstrating that such elements can confer oncogenic c-myc expression. However, in many B-cell lymphomas, Igh-c-myc translocations delete iEmu and place c-myc up to 200 kb upstream of the Igh3'RR. Here we address the oncogenic role of the Igh3'RR by inactivating it in two distinct mouse models for B-cell lymphoma with Igh-c-myc translocations. We show that the Igh3'RR is dispensable for pro-B-cell lymphomas with V(D)J recombination-initiated translocations, but is required for peripheral B-cell lymphomas with CSR-associated translocations. As the Igh3'RR is not required for CSR-associated Igh breaks or Igh-c-myc translocations in peripheral B-cell lymphoma progenitors, we conclude that this regulatory region confers oncogenic activity by long-range and developmental stage-specific activation of translocated c-myc genes.


Subject(s)
3' Untranslated Regions/genetics , Gene Rearrangement, B-Lymphocyte/genetics , Genes, Immunoglobulin Heavy Chain/genetics , Genes, myc/genetics , Lymphoma, B-Cell/genetics , Regulatory Sequences, Nucleic Acid/genetics , Translocation, Genetic/genetics , Alleles , Animals , Cells, Cultured , Chromosome Breakpoints , Immunoglobulin Class Switching/genetics , Lymphoma, B-Cell/pathology , Mice , Mice, Transgenic
12.
Nature ; 460(7252): 231-6, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19587764

ABSTRACT

Variable, diversity and joining gene segment (V(D)J) recombination assembles immunoglobulin heavy or light chain (IgH or IgL) variable region exons in developing bone marrow B cells, whereas class switch recombination (CSR) exchanges IgH constant region exons in peripheral B cells. Both processes use directed DNA double-strand breaks (DSBs) repaired by non-homologous end-joining (NHEJ). Errors in either V(D)J recombination or CSR can initiate chromosomal translocations, including oncogenic IgH locus (Igh) to c-myc (also known as Myc) translocations of peripheral B cell lymphomas. Collaboration between these processes has also been proposed to initiate translocations. However, the occurrence of V(D)J recombination in peripheral B cells is controversial. Here we show that activated NHEJ-deficient splenic B cells accumulate V(D)J-recombination-associated breaks at the lambda IgL locus (Igl), as well as CSR-associated Igh breaks, often in the same cell. Moreover, Igl and Igh breaks are frequently joined to form translocations, a phenomenon associated with specific Igh-Igl co-localization. Igh and c-myc also co-localize in these cells; correspondingly, the introduction of frequent c-myc DSBs robustly promotes Igh-c-myc translocations. Our studies show peripheral B cells that attempt secondary V(D)J recombination, and determine a role for mechanistic factors in promoting recurrent translocations in tumours.


Subject(s)
B-Lymphocytes/metabolism , Gene Rearrangement, B-Lymphocyte/genetics , Genes, Immunoglobulin/genetics , Immunoglobulin Class Switching/genetics , Translocation, Genetic/genetics , Animals , Cytidine Deaminase/deficiency , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , DNA Breaks, Double-Stranded , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Female , Genes, myc/genetics , Homeodomain Proteins/metabolism , Immunoglobulin Heavy Chains/genetics , Immunoglobulin kappa-Chains/genetics , Immunoglobulin lambda-Chains/genetics , Integrases/genetics , Integrases/metabolism , Interphase , Lymphocyte Activation , Male , Mice , Receptors, Complement 3d/genetics , Recombination, Genetic/genetics , Spleen/cytology , Spleen/immunology
13.
J Exp Med ; 205(13): 3079-90, 2008 Dec 22.
Article in English | MEDLINE | ID: mdl-19064702

ABSTRACT

Nonhomologous end-joining (NHEJ) repairs DNA double-strand breaks (DSBs) during V(D)J recombination in developing lymphocytes and during immunoglobulin (Ig) heavy chain (IgH) class switch recombination (CSR) in peripheral B lymphocytes. We now show that CD21-cre-mediated deletion of the Xrcc4 NHEJ gene in p53-deficient peripheral B cells leads to recurrent surface Ig-negative B lymphomas ("CXP lymphomas"). Remarkably, CXP lymphomas arise from peripheral B cells that had attempted both receptor editing (secondary V[D]J recombination of Igkappa and Iglambda light chain genes) and IgH CSR subsequent to Xrcc4 deletion. Correspondingly, CXP tumors frequently harbored a CSR-based reciprocal chromosomal translocation that fused IgH to c-myc, as well as large chromosomal deletions or translocations involving Igkappa or Iglambda, with the latter fusing Iglambda to oncogenes or to IgH. Our findings reveal peripheral B cells that have undergone both editing and CSR and show them to be common progenitors of CXP tumors. Our studies also reveal developmental stage-specific mechanisms of c-myc activation via IgH locus translocations. Thus, Xrcc4/p53-deficient pro-B lymphomas routinely activate c-myc by gene amplification, whereas Xrcc4/p53-deficient peripheral B cell lymphomas routinely ectopically activate a single c-myc copy.


Subject(s)
B-Lymphocytes , Cell Transformation, Neoplastic/immunology , DNA-Binding Proteins/immunology , Gene Rearrangement, B-Lymphocyte , Immunoglobulin Class Switching , Recombination, Genetic , Amino Acid Sequence , Animals , B-Lymphocytes/immunology , B-Lymphocytes/physiology , Base Sequence , DNA Damage , DNA Repair , DNA-Binding Proteins/genetics , Genes, Immunoglobulin Heavy Chain , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Lymphoma/genetics , Lymphoma/immunology , Lymphoma/pathology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Mice , Mice, Knockout , Molecular Sequence Data , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/immunology , Sequence Alignment , Translocation, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology
14.
Mol Cell ; 28(6): 1045-57, 2007 Dec 28.
Article in English | MEDLINE | ID: mdl-18158901

ABSTRACT

Phosphorylated histone H2AX ("gamma-H2AX") recruits MDC1, 53BP1, and BRCA1 to chromatin near a double-strand break (DSB) and facilitates efficient repair of the break. It is unclear to what extent gamma-H2AX-associated proteins act in concert and to what extent their functions within gamma-H2AX chromatin are distinct. We addressed this question by comparing the mechanisms of action of MDC1 and 53BP1 in DSB repair (DSBR). We find that MDC1 functions primarily in homologous recombination/sister chromatid recombination, in a manner strictly dependent upon its ability to interact with gamma-H2AX but, unexpectedly, not requiring recruitment of 53BP1 or BRCA1 to gamma-H2AX chromatin. In contrast, 53BP1 functions in XRCC4-dependent nonhomologous end-joining, likely mediated by its interaction with dimethylated lysine 20 of histone H4 but, surprisingly, independent of H2AX. These results suggest a specialized adaptation of the "histone code" in which distinct histone tail-protein interactions promote engagement of distinct DSBR pathways.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Intracellular Signaling Peptides and Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , BRCA1 Protein/physiology , Blotting, Western , Cell Cycle Proteins , Cell Line , Chromatids/genetics , Chromosomal Proteins, Non-Histone , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Histones/genetics , Histones/metabolism , Histones/physiology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Mice , Microscopy, Fluorescence , Mutation , Protein Binding/radiation effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombination, Genetic/radiation effects , Transfection , Tumor Suppressor p53-Binding Protein 1
15.
Nature ; 449(7161): 478-82, 2007 Sep 27.
Article in English | MEDLINE | ID: mdl-17713479

ABSTRACT

Immunoglobulin variable region exons are assembled in developing B cells by V(D)J recombination. Once mature, these cells undergo class-switch recombination (CSR) when activated by antigen. CSR changes the heavy chain constant region exons (Ch) expressed with a given variable region exon from Cmu to a downstream Ch (for example, Cgamma, Cepsilon or Calpha), thereby switching expression from IgM to IgG, IgE or IgA. Both V(D)J recombination and CSR involve the introduction of DNA double-strand breaks and their repair by means of end joining. For CSR, double-strand breaks are introduced into switch regions that flank Cmu and a downstream Ch, followed by fusion of the broken switch regions. In mammalian cells, the 'classical' non-homologous end joining (C-NHEJ) pathway repairs both general DNA double-strand breaks and programmed double-strand breaks generated by V(D)J recombination. C-NHEJ, as observed during V(D)J recombination, joins ends that lack homology to form 'direct' joins, and also joins ends with several base-pair homologies to form microhomology joins. CSR joins also display direct and microhomology joins, and CSR has been suggested to use C-NHEJ. Xrcc4 and DNA ligase IV (Lig4), which cooperatively catalyse the ligation step of C-NHEJ, are the most specific C-NHEJ factors; they are absolutely required for V(D)J recombination and have no known functions other than C-NHEJ. Here we assess whether C-NHEJ is also critical for CSR by assaying CSR in Xrcc4- or Lig4-deficient mouse B cells. C-NHEJ indeed catalyses CSR joins, because C-NHEJ-deficient B cells had decreased CSR and substantial levels of IgH locus (immunoglobulin heavy chain, encoded by Igh) chromosomal breaks. However, an alternative end-joining pathway, which is markedly biased towards microhomology joins, supports CSR at unexpectedly robust levels in C-NHEJ-deficient B cells. In the absence of C-NHEJ, this alternative end-joining pathway also frequently joins Igh locus breaks to other chromosomes to generate translocations.


Subject(s)
Genes, Immunoglobulin Heavy Chain/genetics , Immunoglobulin Class Switching/genetics , Recombination, Genetic/genetics , Translocation, Genetic/genetics , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , B-Lymphocytes/radiation effects , Base Sequence , Cell Proliferation , Cells, Cultured , Chromosome Breakage , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , In Situ Hybridization, Fluorescence , Mice , Radiation, Ionizing , Telomere/genetics
16.
Adv Immunol ; 94: 157-214, 2007.
Article in English | MEDLINE | ID: mdl-17560275

ABSTRACT

To mount an optimum immune response, mature B lymphocytes can change the class of expressed antibody from IgM to IgG, IgA, or IgE through a recombination/deletion process termed immunoglobulin heavy chain (IgH) class switch recombination (CSR). CSR requires the activation-induced cytidine deaminase (AID), which has been shown to employ single-stranded DNA as a substrate in vitro. IgH CSR occurs within and requires large, repetitive sequences, termed S regions, which are parts of germ line transcription units (termed "C(H) genes") that are composed of promoters, S regions, and individual IgH constant region exons. CSR requires and is directed by germ line transcription of participating C(H) genes prior to CSR. AID deamination of cytidines in S regions appears to lead to S region double-stranded breaks (DSBs) required to initiate CSR. Joining of two broken S regions to complete CSR exploits the activities of general DNA DSB repair mechanisms. In this chapter, we discuss our current knowledge of the function of S regions, germ line transcription, AID, and DNA repair in CSR. We present a model for CSR in which transcription through S regions provides DNA substrates on which AID can generate DSB-inducing lesions. We also discuss how phosphorylation of AID may mediate interactions with cofactors that facilitate access to transcribed S regions during CSR and transcribed variable regions during the related process of somatic hypermutation (SHM). Finally, in the context of this CSR model, we further discuss current findings that suggest synapsis and joining of S region DSBs during CSR have evolved to exploit general mechanisms that function to join widely separated chromosomal DSBs.


Subject(s)
Cytidine Deaminase/physiology , Immunoglobulin Class Switching/genetics , Immunoglobulin Heavy Chains/genetics , Somatic Hypermutation, Immunoglobulin/genetics , Animals , B-Lymphocytes/immunology , Biological Evolution , DNA Breaks, Double-Stranded , DNA Repair , Humans
17.
Proc Natl Acad Sci U S A ; 103(19): 7378-83, 2006 May 09.
Article in English | MEDLINE | ID: mdl-16670198

ABSTRACT

Inactivation of the XRCC4 nonhomologous end-joining factor in the mouse germ line leads to embryonic lethality, in association with apoptosis of newly generated, postmitotic neurons. We now show that conditional inactivation of the XRCC4 in nestin-expressing neuronal progenitor cells, although leading to no obvious phenotype in a WT background, leads to early onset of neuronally differentiated medulloblastomas (MBs) in a p53-deficient background. A substantial proportion of the XRCC4/p53-deficient MBs have high-level N-myc gene amplification, often intrachromosomally in the context of complex translocations or other alterations of chromosome 12, on which N-myc resides, or extrachromosomally within double minutes. In addition, most XRCC4/p53-deficient MBs harbor clonal translocations of chromosome 13, which frequently involve chromosome 6 as a partner. One copy of the patched gene (Ptc), which lies on chromosome 13, was deleted in all tested XRCC4/p53-deficient MBs in the context of translocations or interstitial deletions. In addition, Cyclin D2, a chromosome 6 gene, was amplified in a subset of tumors. Notably, amplification of Myc-family or Cyclin D2 genes and deletion of Ptc also have been observed in human MBs. We therefore conclude that, in neuronal cells of mice, the nonhomologous end-joining pathway plays a critical role in suppressing genomic instability that, in a p53-deficient background, routinely contributes to genesis of MBs with recurrent chromosomal alterations.


Subject(s)
DNA-Binding Proteins/metabolism , Medulloblastoma/metabolism , Translocation, Genetic/genetics , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/metabolism , Alleles , Animals , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Down-Regulation/genetics , Gene Amplification , Intermediate Filament Proteins/metabolism , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Nestin , Survival Rate , Time Factors , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
18.
Mol Cell ; 21(2): 201-14, 2006 Jan 20.
Article in English | MEDLINE | ID: mdl-16427010

ABSTRACT

Histone H2AX promotes DNA double-strand break (DSB) repair and immunoglobulin heavy chain (IgH) class switch recombination (CSR) in B-lymphocytes. CSR requires activation-induced cytidine deaminase (AID) and involves joining of DSB intermediates by end joining. We find that AID-dependent IgH locus chromosome breaks occur at high frequency in primary H2AX-deficient B cells activated for CSR and that a substantial proportion of these breaks participate in chromosomal translocations. Moreover, activated B cells deficient for ATM, 53BP1, or MDC1, which interact with H2AX during the DSB response, show similarly increased IgH locus breaks and translocations. Thus, our findings implicate a general role for these factors in promoting end joining and thereby preventing DSBs from progressing into chromosomal breaks and translocations. As cellular p53 status does not markedly influence the frequency of such events, our results also have implications for how p53 and the DSB response machinery cooperate to suppress generation of lymphomas with oncogenic translocations.


Subject(s)
DNA Damage , DNA Repair/physiology , Histones/metabolism , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Chromosome Breakage , Cytidine Deaminase/metabolism , Histones/deficiency , Histones/genetics , Immunoglobulin Class Switching , Immunoglobulin Heavy Chains/genetics , In Situ Hybridization, Fluorescence , In Vitro Techniques , Mice , Mice, Knockout , Translocation, Genetic , Tumor Suppressor Protein p53/metabolism
19.
EMBO J ; 22(14): 3602-12, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12853475

ABSTRACT

The Wiskott-Aldrich syndrome related protein WAVE2 is implicated in the regulation of actin-cytoskeletal reorganization downstream of the small Rho GTPase, Rac. We inactivated the WAVE2 gene by gene-targeted mutation to examine its role in murine development and in actin assembly. WAVE2-deficient embryos survived until approximately embryonic day 12.5 and displayed growth retardation and certain morphological defects, including malformations of the ventricles in the developing brain. WAVE2-deficient embryonic stem cells displayed normal proliferation, whereas WAVE2-deficient embryonic fibroblasts exhibited severe growth defects, as well as defective cell motility in response to PDGF, lamellipodium formation and Rac-mediated actin polymerization. These results imply a non-redundant role for WAVE2 in murine embryogenesis and a critical role for WAVE2 in actin-based processes downstream of Rac that are essential for cell movement.


Subject(s)
Actins/metabolism , Cell Movement/physiology , Embryo, Mammalian/physiology , Microfilament Proteins/deficiency , Microfilament Proteins/metabolism , rac GTP-Binding Proteins/metabolism , Actins/physiology , Animals , Biopolymers , Cell Line , Cytoskeleton/metabolism , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Deletion , Mice , Mice, Knockout , Microfilament Proteins/genetics , Mutation , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/metabolism , Pseudopodia/metabolism , RNA/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Wiskott-Aldrich Syndrome/blood , Wiskott-Aldrich Syndrome/metabolism , Wiskott-Aldrich Syndrome Protein Family
20.
Proc Natl Acad Sci U S A ; 99(12): 8173-8, 2002 Jun 11.
Article in English | MEDLINE | ID: mdl-12034884

ABSTRACT

In mammalian cells, DNA double-strand breaks (DSBs) cause rapid phosphorylation of the H2AX core histone variant (to form gamma-H2AX) in megabase chromatin domains flanking sites of DNA damage. To investigate the role of H2AX in mammalian cells, we generated H2AX-deficient (H2AX(Delta)/Delta) mouse embryonic stem (ES) cells. H2AX(Delta)/Delta ES cells are viable. However, they are highly sensitive to ionizing radiation (IR) and exhibit elevated levels of spontaneous and IR-induced genomic instability. Notably, H2AX is not required for NHEJ per se because H2AX(Delta)/Delta ES cells support normal levels and fidelity of V(D)J recombination in transient assays and also support lymphocyte development in vivo. However, H2AX(Delta)/Delta ES cells exhibit altered IR-induced BRCA1 focus formation. Our findings indicate that H2AX function is essential for mammalian DNA repair and genomic stability.


Subject(s)
Histones/radiation effects , Stem Cells/radiation effects , Amino Acid Sequence , Animals , Antibodies , BRCA1 Protein/genetics , Base Sequence , Blotting, Western , Chromatin/genetics , DNA Damage , DNA Primers , Genes, BRCA1 , Genetic Variation , Histones/deficiency , Histones/genetics , Mice , Mice, Knockout , Oligopeptides/chemistry , Oligopeptides/immunology , Phosphorylation , Polymerase Chain Reaction , Radiation, Ionizing , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...