Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
1.
Annu Rev Immunol ; 30: 175-202, 2012.
Article in English | MEDLINE | ID: mdl-22224778

ABSTRACT

Developing lymphocytes must assemble antigen receptor genes encoding the B cell and T cell receptors. This process is executed by the V(D)J recombination reaction, which can be divided into DNA cleavage and DNA joining steps. The former is carried out by a lymphocyte-specific RAG endonuclease, which mediates DNA cleavage at two recombining gene segments and their flanking RAG recognition sequences. RAG cleavage generates four broken DNA ends that are repaired by nonhomologous end joining forming coding and signal joints. On rare occasions, these DNA ends may join aberrantly forming chromosomal lesions such as translocations, deletions and inversions that have the potential to cause cellular transformation and lymphoid tumors. We discuss the activation of DNA damage responses by RAG-induced DSBs focusing on the component pathways that promote their normal repair and guard against their aberrant resolution. Moreover, we discuss how this DNA damage response impacts processes important for lymphocyte development.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , V(D)J Recombination , Animals , Genomic Instability , Humans , Lymphocytes/immunology , Lymphocytes/metabolism , Receptors, Antigen/genetics
2.
Genes Dev ; 35(19-20): 1356-1367, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34503990

ABSTRACT

Double-strand break (DSB) repair choice is greatly influenced by the initial processing of DNA ends. 53BP1 limits the formation of recombinogenic single-strand DNA (ssDNA) in BRCA1-deficient cells, leading to defects in homologous recombination (HR). However, the exact mechanisms by which 53BP1 inhibits DSB resection remain unclear. Previous studies have identified two potential pathways: protection against DNA2/EXO1 exonucleases presumably through the Shieldin (SHLD) complex binding to ssDNA, and localized DNA synthesis through the CTC1-STN1-TEN1 (CST) and DNA polymerase α (Polα) to counteract resection. Using a combinatorial approach of END-seq, SAR-seq, and RPA ChIP-seq, we directly assessed the extent of resection, DNA synthesis, and ssDNA, respectively, at restriction enzyme-induced DSBs. We show that, in the presence of 53BP1, Polα-dependent DNA synthesis reduces the fraction of resected DSBs and the resection lengths in G0/G1, supporting a previous model that fill-in synthesis can limit the extent of resection. However, in the absence of 53BP1, Polα activity is sustained on ssDNA yet does not substantially counter resection. In contrast, EXO1 nuclease activity is essential for hyperresection in the absence of 53BP1. Thus, Polα-mediated fill-in partially limits resection in the presence of 53BP1 but cannot counter extensive hyperresection due to the loss of 53BP1 exonuclease blockade. These data provide the first nucleotide mapping of DNA synthesis at resected DSBs and provide insight into the relationship between fill-in polymerases and resection exonucleases.


Subject(s)
DNA Breaks, Double-Stranded , DNA Replication , DNA Repair/genetics , DNA Replication/genetics , DNA, Single-Stranded/genetics , Homologous Recombination/genetics , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism
3.
Cell ; 153(6): 1266-80, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23727112

ABSTRACT

The DNA damage response (DDR) protein 53BP1 protects DNA ends from excessive resection in G1, and thereby favors repair by nonhomologous end-joining (NHEJ) as opposed to homologous recombination (HR). During S phase, BRCA1 antagonizes 53BP1 to promote HR. The pro-NHEJ and antirecombinase functions of 53BP1 are mediated in part by RIF1, the only known factor that requires 53BP1 phosphorylation for its recruitment to double-strand breaks (DSBs). Here, we show that a 53BP1 phosphomutant, 53BP18A, comprising alanine substitutions of the eight most N-terminal S/TQ phosphorylation sites, mimics 53BP1 deficiency by restoring genome stability in BRCA1-deficient cells yet behaves like wild-type 53BP1 with respect to immunoglobulin class switch recombination (CSR). 53BP18A recruits RIF1 but fails to recruit the DDR protein PTIP to DSBs, and disruption of PTIP phenocopies 53BP18A. We conclude that 53BP1 promotes productive CSR and suppresses mutagenic DNA repair through distinct phosphodependent interactions with RIF1 and PTIP.


Subject(s)
Carrier Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA End-Joining Repair , DNA-Binding Proteins/metabolism , Immunoglobulin Class Switching , Nuclear Proteins/metabolism , Telomere-Binding Proteins/metabolism , Animals , B-Lymphocytes/metabolism , BRCA1 Protein/metabolism , Chromosomal Proteins, Non-Histone/genetics , DNA Breaks, Double-Stranded , DNA-Binding Proteins/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Genomic Instability , Mice , Mutation , Tumor Suppressor p53-Binding Protein 1
4.
Mol Cell ; 71(2): 332-342.e8, 2018 07 19.
Article in English | MEDLINE | ID: mdl-30017584

ABSTRACT

The modulator of retrovirus infection (MRI or CYREN) is a 30-kDa protein with a conserved N-terminal Ku-binding motif (KBM) and a C-terminal XLF-like motif (XLM). We show that MRI is intrinsically disordered and interacts with many DNA damage response (DDR) proteins, including the kinases ataxia telangiectasia mutated (ATM) and DNA-PKcs and the classical non-homologous end joining (cNHEJ) factors Ku70, Ku80, XRCC4, XLF, PAXX, and XRCC4. MRI forms large multimeric complexes that depend on its N and C termini and localizes to DNA double-strand breaks (DSBs), where it promotes the retention of DDR factors. Mice deficient in MRI and XLF exhibit embryonic lethality at a stage similar to those deficient in the core cNHEJ factors XRCC4 or DNA ligase IV. Moreover, MRI is required for cNHEJ-mediated DSB repair in XLF-deficient lymphocytes. We propose that MRI is an adaptor that, through multivalent interactions, increases the avidity of DDR factors to DSB-associated chromatin to promote cNHEJ.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , Animals , Cell Cycle Proteins/metabolism , Chromatin/genetics , Chromatin/metabolism , DNA Ligase ATP/genetics , DNA Repair , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Ku Autoantigen/genetics , Mice
5.
Nat Immunol ; 13(11): 1092-100, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23001146

ABSTRACT

Germinal centers (GCs) are sites of intense B cell proliferation and are central for T cell-dependent antibody responses. However, the role of c-Myc, a key cell-cycle regulator, in this process has been questioned. Here we identified c-Myc(+) B cell subpopulations in immature and mature GCs and found, by genetic ablation of Myc, that they had indispensable roles in the formation and maintenance of GCs. The identification of these functionally critical cellular subsets has implications for human B cell lymphomagenesis, which originates mostly from GC B cells and frequently involves MYC chromosomal translocations. As these translocations are generally dependent on transcription of the recombining partner loci, the c-Myc(+) GC subpopulations may be at a particularly high risk for malignant transformation.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocytes/metabolism , Cell Cycle/genetics , Germinal Center/metabolism , Proto-Oncogene Proteins c-myc/genetics , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Cycle/immunology , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Gene Deletion , Gene Expression Regulation/immunology , Genes, Reporter , Genetic Loci , Germinal Center/immunology , Germinal Center/pathology , Green Fluorescent Proteins , Lymphoma/genetics , Lymphoma/metabolism , Lymphoma/pathology , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-myc/deficiency , Proto-Oncogene Proteins c-myc/immunology , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Translocation, Genetic
6.
Mol Cell ; 63(5): 898-911, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27477910

ABSTRACT

DNA double-strand breaks (DSBs) arise during physiological transcription, DNA replication, and antigen receptor diversification. Mistargeting or misprocessing of DSBs can result in pathological structural variation and mutation. Here we describe a sensitive method (END-seq) to monitor DNA end resection and DSBs genome-wide at base-pair resolution in vivo. We utilized END-seq to determine the frequency and spectrum of restriction-enzyme-, zinc-finger-nuclease-, and RAG-induced DSBs. Beyond sequence preference, chromatin features dictate the repertoire of these genome-modifying enzymes. END-seq can detect at least one DSB per cell among 10,000 cells not harboring DSBs, and we estimate that up to one out of 60 cells contains off-target RAG cleavage. In addition to site-specific cleavage, we detect DSBs distributed over extended regions during immunoglobulin class-switch recombination. Thus, END-seq provides a snapshot of DNA ends genome-wide, which can be utilized for understanding genome-editing specificities and the influence of chromatin on DSB pathway choice.


Subject(s)
Chromatin/chemistry , DNA Breaks, Double-Stranded , DNA/genetics , Genome , High-Throughput Nucleotide Sequencing/methods , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Chromatin/immunology , DNA/immunology , DNA Replication , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Gene Expression Regulation , Histones/genetics , Histones/immunology , Immunoglobulin Class Switching/genetics , Mice , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Recombination, Genetic , Thymocytes/cytology , Thymocytes/immunology
7.
Cell ; 135(6): 1009-12, 2008 Dec 12.
Article in English | MEDLINE | ID: mdl-19070571

ABSTRACT

Chromosomal translocations that juxtapose antigen receptor genes and oncogenes are frequently associated with lymphoid malignancies. In this issue, Robbiani et al. (2008) show that activation-induced deaminase (AID), an enzyme involved in antigen receptor gene diversification, generates DNA double-strand breaks (DSBs) in oncogenes, and Tsai et al. (2008) propose that AID and the recombinase-activating gene (RAG) endonuclease may collaborate to generate off-target DSBs.


Subject(s)
Cytidine Deaminase/metabolism , DNA Breaks, Double-Stranded , Receptors, Antigen/genetics , Animals , DNA Repair , Homeodomain Proteins/metabolism , Humans , Lymphoma/genetics , Lymphoma/metabolism , Receptors, Antigen/metabolism , Translocation, Genetic
8.
Nat Immunol ; 11(3): 207-15, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20081848

ABSTRACT

Hematopoietic stem cell (HSC) differentiation is regulated by cell-intrinsic and cell-extrinsic cues. In addition to transcriptional regulation, post-translational regulation may also control HSC differentiation. To test this hypothesis, we visualized the ubiquitin-regulated protein stability of a single transcription factor, c-Myc. The stability of c-Myc protein was indicative of HSC quiescence, and c-Myc protein abundance was controlled by the ubiquitin ligase Fbw7. Fine changes in the stability of c-Myc protein regulated the HSC gene-expression signature. Using whole-genome genomic approaches, we identified specific regulators of HSC function directly controlled by c-Myc binding; however, adult HSCs and embryonic stem cells sensed and interpreted c-Myc-regulated gene expression in distinct ways. Our studies show that a ubiquitin ligase-substrate pair can orchestrate the molecular program of HSC differentiation.


Subject(s)
Cell Differentiation/physiology , Hematopoietic Stem Cells/cytology , Ubiquitin-Protein Ligases/immunology , Animals , Cell Cycle/genetics , Cell Cycle/immunology , Cell Cycle Proteins/immunology , Cell Differentiation/genetics , Chromatin Immunoprecipitation , Flow Cytometry , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Mice , Mice, Knockout , Proto-Oncogene Proteins c-myc/immunology
9.
Mol Cell ; 54(6): 1022-1033, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24837676

ABSTRACT

The carboxy-terminal binding protein (CtBP)-interacting protein (CtIP) is known to function in 5' strand resection during homologous recombination, similar to the budding yeast Sae2 protein, but its role in this process is unclear. Here, we characterize recombinant human CtIP and find that it exhibits 5' flap endonuclease activity on branched DNA structures, independent of the MRN complex. Phosphorylation of CtIP at known damage-dependent sites and other sites is essential for its catalytic activity, although the S327 and T847 phosphorylation sites are dispensable. A catalytic mutant of CtIP that is deficient in endonuclease activity exhibits wild-type levels of homologous recombination at restriction enzyme-generated breaks but is deficient in processing topoisomerase adducts and radiation-induced breaks in human cells, suggesting that the nuclease activity of CtIP is specifically required for the removal of DNA adducts at sites of DNA breaks.


Subject(s)
Carrier Proteins/metabolism , DNA Breaks, Double-Stranded , DNA End-Joining Repair/genetics , Endonucleases/metabolism , Nuclear Proteins/metabolism , Recombinational DNA Repair/genetics , Binding Sites/genetics , Carrier Proteins/genetics , Catalysis , Cell Line , Cell Survival/genetics , DNA/genetics , DNA-Binding Proteins/genetics , Endodeoxyribonucleases , Endonucleases/genetics , Humans , Nuclear Proteins/genetics , Phosphorylation/genetics , Protein Processing, Post-Translational/genetics , Radiation, Ionizing , Recombination, Genetic
10.
Mol Cell ; 56(6): 808-18, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25435138

ABSTRACT

The structure of broken DNA ends is a critical determinant of the pathway used for DNA double-strand break (DSB) repair. Here, we develop an approach involving the hairpin capture of DNA end structures (HCoDES), which elucidates chromosomal DNA end structures at single-nucleotide resolution. HCoDES defines structures of physiologic DSBs generated by the RAG endonuclease, as well as those generated by nucleases widely used for genome editing. Analysis of G1 phase cells deficient in H2AX or 53BP1 reveals DNA ends that are frequently resected to form long single-stranded overhangs that can be repaired by mutagenic pathways. In addition to 3' overhangs, many of these DNA ends unexpectedly form long 5' single-stranded overhangs. The divergence in DNA end structures resolved by HCoDES suggests that H2AX and 53BP1 may have distinct activities in end protection. Thus, the high-resolution end structures obtained by HCoDES identify features of DNA end processing during DSB repair.


Subject(s)
Chromosomes, Human/genetics , Sequence Analysis, DNA/methods , Base Sequence , Cells, Cultured , DNA Breaks, Double-Stranded , Humans
11.
Nat Immunol ; 10(6): 655-64, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19448632

ABSTRACT

Coordinated recombination of homologous antigen receptor loci is thought to be important for allelic exclusion. Here we show that homologous immunoglobulin alleles pair in a stage-specific way that mirrors the recombination patterns of these loci. The frequency of homologous immunoglobulin pairing was much lower in the absence of the RAG-1-RAG-2 recombinase and was restored in Rag1-/- developing B cells with a transgene expressing a RAG-1 active-site mutant that supported DNA binding but not cleavage. The introduction of DNA breaks on one immunoglobulin allele induced ATM-dependent repositioning of the other allele to pericentromeric heterochromatin. ATM activated by the cleaved allele acts in trans on the uncleaved allele to prevent biallelic recombination and chromosome breaks or translocations.


Subject(s)
Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Immunoglobulins/genetics , Protein Serine-Threonine Kinases/genetics , Recombination, Genetic , Tumor Suppressor Proteins/genetics , Alleles , Animals , Ataxia Telangiectasia Mutated Proteins , B-Lymphocytes/metabolism , Cells, Cultured , DNA Breaks , Gene Rearrangement , Mice , Mice, Inbred C57BL , Mice, Knockout , VDJ Recombinases/metabolism
12.
Nature ; 507(7491): 243-7, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24509714

ABSTRACT

The transcription factors c-Myc and N-Myc--encoded by Myc and Mycn, respectively--regulate cellular growth and are required for embryonic development. A third paralogue, Mycl1, is dispensable for normal embryonic development but its biological function has remained unclear. To examine the in vivo function of Mycl1 in mice, we generated an inactivating Mycl1(gfp) allele that also reports Mycl1 expression. We find that Mycl1 is selectively expressed in dendritic cells (DCs) of the immune system and controlled by IRF8, and that during DC development, Mycl1 expression is initiated in the common DC progenitor concurrent with reduction in c-Myc expression. Mature DCs lack expression of c-Myc and N-Myc but maintain L-Myc expression even in the presence of inflammatory signals such as granulocyte-macrophage colony-stimulating factor. All DC subsets develop in Mycl1-deficient mice, but some subsets such as migratory CD103(+) conventional DCs in the lung and liver are greatly reduced at steady state. Importantly, loss of L-Myc by DCs causes a significant decrease in in vivo T-cell priming during infection by Listeria monocytogenes and vesicular stomatitis virus. The replacement of c-Myc by L-Myc in immature DCs may provide for Myc transcriptional activity in the setting of inflammation that is required for optimal T-cell priming.


Subject(s)
Cross-Priming/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation , Proto-Oncogene Proteins c-myc/metabolism , T-Lymphocytes/immunology , Animals , Antigens, CD/metabolism , Cell Division , Dendritic Cells/cytology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Inflammation/immunology , Inflammation/metabolism , Integrin alpha Chains/metabolism , Interferon Regulatory Factors/metabolism , Listeria monocytogenes/immunology , Liver/cytology , Liver/immunology , Lung/cytology , Lung/immunology , Male , Mice , Proto-Oncogene Proteins c-myc/deficiency , Transcription, Genetic , Vesiculovirus/immunology
13.
Blood ; 128(6): 783-93, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27301863

ABSTRACT

Primary immunodeficiency diseases comprise a group of heterogeneous genetic defects that affect immune system development and/or function. Here we use in vitro differentiation of human induced pluripotent stem cells (iPSCs) generated from patients with different recombination-activating gene 1 (RAG1) mutations to assess T-cell development and T-cell receptor (TCR) V(D)J recombination. RAG1-mutants from severe combined immunodeficient (SCID) patient cells showed a failure to sustain progression beyond the CD3(--)CD4(-)CD8(-)CD7(+)CD5(+)CD38(-)CD31(-/lo)CD45RA(+) stage of T-cell development to reach the CD3(-/+)CD4(+)CD8(+)CD7(+)CD5(+)CD38(+)CD31(+)CD45RA(-) stage. Despite residual mutant RAG1 recombination activity from an Omenn syndrome (OS) patient, similar impaired T-cell differentiation was observed, due to increased single-strand DNA breaks that likely occur due to heterodimers consisting of both an N-terminal truncated and a catalytically dead RAG1. Furthermore, deep-sequencing analysis of TCR-ß (TRB) and TCR-α (TRA) rearrangements of CD3(-)CD4(+)CD8(-) immature single-positive and CD3(+)CD4(+)CD8(+) double-positive cells showed severe restriction of repertoire diversity with preferential usage of few Variable, Diversity, and Joining genes, and skewed length distribution of the TRB and TRA complementary determining region 3 sequences from SCID and OS iPSC-derived cells, whereas control iPSCs yielded T-cell progenitors with a broadly diversified repertoire. Finally, no TRA/δ excision circles (TRECs), a marker of TRA/δ locus rearrangements, were detected in SCID and OS-derived T-lineage cells, consistent with a pre-TCR block in T-cell development. This study compares human T-cell development of SCID vs OS patients, and elucidates important differences that help to explain the wide range of immunologic phenotypes that result from different mutations within the same gene of various patients.


Subject(s)
Homeodomain Proteins/genetics , Induced Pluripotent Stem Cells/pathology , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/pathology , T-Lymphocytes/pathology , Cells, Cultured , DNA Breaks , Genes, RAG-1 , Humans , Infant , Mutation , Receptors, Antigen, T-Cell, alpha-beta/genetics , V(D)J Recombination
14.
Nature ; 469(7329): 245-9, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-21160476

ABSTRACT

DNA double-strand breaks (DSBs) are generated by the recombination activating gene (RAG) endonuclease in all developing lymphocytes as they assemble antigen receptor genes. DNA cleavage by RAG occurs only at the G1 phase of the cell cycle and generates two hairpin-sealed DNA (coding) ends that require nucleolytic opening before their repair by classical non-homologous end-joining (NHEJ). Although there are several cellular nucleases that could perform this function, only the Artemis nuclease is able to do so efficiently. Here, in vivo, we show that in murine cells the histone protein H2AX prevents nucleases other than Artemis from processing hairpin-sealed coding ends; in the absence of H2AX, CtIP can efficiently promote the hairpin opening and resection of DNA ends generated by RAG cleavage. This CtIP-mediated resection is inhibited by γ-H2AX and by MDC-1 (mediator of DNA damage checkpoint 1), which binds to γ-H2AX in chromatin flanking DNA DSBs. Moreover, the ataxia telangiectasia mutated (ATM) kinase activates antagonistic pathways that modulate this resection. CtIP DNA end resection activity is normally limited to cells at post-replicative stages of the cell cycle, in which it is essential for homology-mediated repair. In G1-phase lymphocytes, DNA ends that are processed by CtIP are not efficiently joined by classical NHEJ and the joints that do form frequently use micro-homologies and show significant chromosomal deletions. Thus, H2AX preserves the structural integrity of broken DNA ends in G1-phase lymphocytes, thereby preventing these DNA ends from accessing repair pathways that promote genomic instability.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Repair , G1 Phase , Gene Rearrangement, B-Lymphocyte , Histones/metabolism , Lymphocytes/metabolism , Adaptor Proteins, Signal Transducing , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Line, Transformed , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Endonucleases , Gene Rearrangement, B-Lymphocyte/genetics , Genomic Instability , Histones/deficiency , Histones/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lymphocytes/cytology , Mice , Nuclear Proteins , Precursor Cells, B-Lymphoid/metabolism , Protein Serine-Threonine Kinases/metabolism , Recombination, Genetic/genetics , Substrate Specificity , Tumor Suppressor Proteins/metabolism
15.
Mol Cell ; 34(3): 298-310, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19450528

ABSTRACT

A hallmark of the cellular response to DNA double-strand breaks (DSBs) is histone H2AX phosphorylation in chromatin to generate gamma-H2AX. Here, we demonstrate that gamma-H2AX densities increase transiently along DNA strands as they are broken and repaired in G1 phase cells. The region across which gamma-H2AX forms does not spread as DSBs persist; rather, gamma-H2AX densities equilibrate at distinct levels within a fixed distance from DNA ends. Although both ATM and DNA-PKcs generate gamma-H2AX, only ATM promotes gamma-H2AX formation to maximal distance and maintains gamma-H2AX densities. MDC1 is essential for gamma-H2AX formation at high densities near DSBs, but not for generation of gamma-H2AX over distal sequences. Reduced H2AX levels in chromatin impair the density, but not the distance, of gamma-H2AX formed. Our data suggest that H2AX fuels a gamma-H2AX self-reinforcing mechanism that retains MDC1 and activated ATM in chromatin near DSBs and promotes continued local phosphorylation of H2AX.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatin/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , DNA/metabolism , Histones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Ataxia Telangiectasia Mutated Proteins , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Endonucleases , G1 Phase/physiology , Genes, T-Cell Receptor alpha/genetics , Histones/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Recombination, Genetic , Thymus Gland/cytology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics
16.
Proc Natl Acad Sci U S A ; 111(1): 391-6, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24371308

ABSTRACT

Deregulated activation of ß-catenin in cancer has been correlated with genomic instability. During thymocyte development, ß-catenin activates transcription in partnership with T-cell-specific transcription factor 1 (Tcf-1). We previously reported that targeted activation of ß-catenin in thymocytes (CAT mice) induces lymphomas that depend on recombination activating gene (RAG) and myelocytomatosis oncogene (Myc) activities. Here we show that these lymphomas have recurring Tcra/Myc translocations that resulted from illegitimate RAG recombination events and resembled oncogenic translocations previously described in human T-ALL. We therefore used the CAT animal model to obtain mechanistic insights into the transformation process. ChIP-seq analysis uncovered a link between Tcf-1 and RAG2 showing that the two proteins shared binding sites marked by trimethylated histone-3 lysine-4 (H3K4me3) throughout the genome, including near the translocation sites. Pretransformed CAT thymocytes had increased DNA damage at the translocating loci and showed altered repair of RAG-induced DNA double strand breaks. These cells were able to survive despite DNA damage because activated ß-catenin promoted an antiapoptosis gene expression profile. Thus, activated ß-catenin promotes genomic instability that leads to T-cell lymphomas as a consequence of altered double strand break repair and increased survival of thymocytes with damaged DNA.


Subject(s)
Genomic Instability , Lymphocyte Activation , Lymphoma/genetics , T-Lymphocytes/cytology , beta Catenin/metabolism , Animals , Apoptosis , Base Sequence , Cell Survival , DNA Breaks, Double-Stranded , DNA Methylation , DNA Repair , Disease Models, Animal , Genes, RAG-1/genetics , Hepatocyte Nuclear Factor 1-alpha , Histones/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Recombination, Genetic , T Cell Transcription Factor 1/metabolism , Thymocytes/cytology , Translocation, Genetic , beta Catenin/genetics
17.
Blood ; 123(25): 3909-13, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24795346

ABSTRACT

Previous studies have established pivotal roles for c-Myc and its homolog N-Myc in hematopoietic stem cell (HSC) maintenance and niche-dependent differentiation. However, it remains largely unclear how c-Myc expression is regulated in this context. Here, we show that HSCs and more committed progenitors express similar levels of c-myc transcripts. Using knock-in mice expressing a functional enhanced green fluorescent protein-c-Myc fusion protein under control of the endogenous c-myc locus, c-Myc protein levels were assessed. Although HSCs express low levels of c-Myc protein, its expression increases steadily during progenitor differentiation. Thus, mRNA and protein expression patterns differ significantly in stem/progenitor cells, suggesting that c-Myc expression is largely controlled posttranscriptionally. Moreover, interferon-α exposure, which activates dormant HSCs, strongly induces c-Myc expression at the protein level but not at the transcript level. This posttranscriptional mechanism of c-Myc regulation provides the blood system with a rapid way to adjust c-Myc expression according to demand during hematopoietic stress.


Subject(s)
Gene Expression Regulation/drug effects , Hematopoietic Stem Cells/drug effects , Homeostasis/drug effects , Interferon-alpha/pharmacology , Proto-Oncogene Proteins c-myc/genetics , RNA Processing, Post-Transcriptional , Animals , Cell Differentiation/genetics , Cells, Cultured , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Homeostasis/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-myc/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stress, Physiological/genetics
18.
Proc Natl Acad Sci U S A ; 110(24): 9873-8, 2013 Jun 11.
Article in English | MEDLINE | ID: mdl-23716691

ABSTRACT

The ability to sense metabolic stress is critical for successful cellular adaptation. In eukaryotes, the AMP-activated protein kinase (AMPK), a highly conserved serine/threonine kinase, functions as a critical metabolic sensor. AMPK is activated by the rising ADP/ATP and AMP/ATP ratios during conditions of energy depletion and also by increasing intracellular Ca(2+). In response to metabolic stress, AMPK maintains energy homeostasis by phosphorylating and regulating proteins that are involved in many physiological processes including glucose and fatty acid metabolism, transcription, cell growth, mitochondrial biogenesis, and autophagy. Evidence is mounting that AMPK also plays a role in a number of pathways unrelated to energy metabolism. Here, we identify the recombination-activating gene 1 protein (RAG1) as a substrate of AMPK. The RAG1/RAG2 complex is a lymphoid-specific endonuclease that catalyzes specific DNA cleavage during V(D)J recombination, which is required for the assembly of the Ig and T-cell receptor genes of the immune system. AMPK directly phosphorylates RAG1 at serine 528, and the phosphorylation enhances the catalytic activity of the RAG complex, resulting in increased cleavage of oligonucleotide substrates in vitro, or increased recombination of an extrachromosomal substrate in a cellular assay. Our results suggest that V(D)J recombination can be regulated by AMPK activation, providing a potential new link between metabolic stress and development of B and T lymphocytes.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Homeodomain Proteins/metabolism , Serine/metabolism , V(D)J Recombination , Amino Acid Sequence , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Immunoblotting , Mice , Mice, Knockout , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotides/genetics , Oligonucleotides/metabolism , Phosphorylation , Sequence Homology, Amino Acid , Serine/genetics , Substrate Specificity
19.
J Allergy Clin Immunol ; 136(1): 140-150.e7, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25917813

ABSTRACT

BACKGROUND: The endonuclease ARTEMIS, which is encoded by the DCLRE1C gene, is a component of the nonhomologous end-joining pathway and participates in hairpin opening during the V(D)J recombination process and repair of a subset of DNA double-strand breaks. Patients with ARTEMIS deficiency usually present with severe combined immunodeficiency (SCID) and cellular radiosensitivity, but hypomorphic mutations can cause milder phenotypes (leaky SCID). OBJECTIVE: We sought to correlate the functional effect of human DCLRE1C mutations on phenotypic presentation in patients with ARTEMIS deficiency. METHODS: We studied the recombination and DNA repair activity of 41 human DCLRE1C mutations in Dclre1c(-/-) v-abl kinase-transformed pro-B cells retrovirally engineered with a construct that allows quantification of recombination activity by means of flow cytometry. For assessment of DNA repair efficacy, resolution of γH2AX accumulation was studied after ionizing radiation. RESULTS: Low or absent activity was detected for mutations causing a typical SCID phenotype. Most of the patients with leaky SCID were compound heterozygous for 1 loss-of-function and 1 hypomorphic allele, with significant residual levels of recombination and DNA repair activity. Deletions disrupting the C-terminus result in truncated but partially functional proteins and are often associated with leaky SCID. Overexpression of hypomorphic mutants might improve the functional defect. CONCLUSIONS: Correlation between the nature and location of DCLRE1C mutations, functional activity, and the clinical phenotype has been observed. Hypomorphic variants that have been reported in the general population can be disease causing if combined in trans with a loss-of-function allele. Therapeutic strategies aimed at inducing overexpression of hypomorphic alleles might be beneficial.


Subject(s)
B-Lymphocytes/physiology , Mutation/genetics , Nuclear Proteins/genetics , Severe Combined Immunodeficiency/genetics , Adolescent , Adult , Alleles , B-Lymphocytes/radiation effects , Cell Line, Transformed , Child , Child, Preschool , DNA Mutational Analysis , DNA Repair/genetics , DNA-Binding Proteins , Endonucleases , Heterozygote , Histones/metabolism , Humans , Infant , Infant, Newborn , Male , Oncogene Proteins v-abl/genetics , Oncogene Proteins v-abl/metabolism , Phenotype , Radiation Tolerance/genetics , Radiation, Ionizing , V(D)J Recombination/genetics , Young Adult
20.
J Immunol ; 190(6): 2485-9, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23420891

ABSTRACT

During early stages of B-lineage differentiation in bone marrow, signals emanating from IL-7R and pre-BCR are thought to synergistically induce proliferative expansion of progenitor cells. Paradoxically, loss of pre-BCR-signaling components is associated with leukemia in both mice and humans. Exactly how progenitor B cells perform the task of balancing proliferative burst dependent on IL-7 with the termination of IL-7 signals and the initiation of L chain gene rearrangement remains to be elucidated. In this article, we provide genetic and functional evidence that the cessation of the IL-7 response of pre-B cells is controlled via a cell-autonomous mechanism that operates at a discrete developmental transition inside Fraction C' (large pre-BII) marked by transient expression of c-Myc. Our data indicate that pre-BCR cooperates with IL-7R in expanding the pre-B cell pool, but it is also critical to control the differentiation program shutting off the c-Myc gene in large pre-B cells.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Down-Regulation/immunology , Interleukin-7/physiology , Proto-Oncogene Proteins c-myc/genetics , Stem Cells/immunology , Stem Cells/metabolism , Animals , B-Lymphocyte Subsets/pathology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Cells, Cultured , Coculture Techniques , Down-Regulation/genetics , Gene Knock-In Techniques , Growth Inhibitors/biosynthesis , Growth Inhibitors/genetics , Interleukin-7/antagonists & inhibitors , Interleukin-7/metabolism , Mice , Mice, Knockout , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-myc/biosynthesis , Receptors, Interleukin-7/antagonists & inhibitors , Receptors, Interleukin-7/physiology , Signal Transduction/genetics , Signal Transduction/immunology , Stem Cells/pathology , Stromal Cells/immunology , Stromal Cells/metabolism , Stromal Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL