Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Exp Med ; 217(9)2020 09 07.
Article in English | MEDLINE | ID: mdl-32526772

ABSTRACT

The monoallelic expression of antigen receptor (AgR) genes, called allelic exclusion, is fundamental for highly specific immune responses to pathogens. This cardinal feature of adaptive immunity is achieved by the assembly of a functional AgR gene on one allele, with subsequent feedback inhibition of V(D)J recombination on the other allele. A range of epigenetic mechanisms have been implicated in sequential recombination of AgR alleles; however, we now demonstrate that a genetic mechanism controls this process for Tcrb. Replacement of V(D)J recombinase targets at two different mouse Vß gene segments with a higher quality target elevates Vß rearrangement frequency before feedback inhibition, dramatically increasing the frequency of T cells with TCRß chains derived from both Tcrb alleles. Thus, TCRß allelic exclusion is enforced genetically by the low quality of Vß recombinase targets that stochastically restrict the production of two functional rearrangements before feedback inhibition silences one allele.


Subject(s)
Alleles , Protein Sorting Signals , Receptors, Antigen, T-Cell, alpha-beta/genetics , V(D)J Recombination/genetics , Animals , Base Sequence , Feedback, Physiological , Gene Expression Regulation , Hybridomas , Mice, Inbred C57BL , Mice, Transgenic , Protein Binding , Proto-Oncogene Proteins c-fos/metabolism , T-Lymphocytes/cytology , Thymocytes/cytology
2.
J Immunol ; 195(3): 1262-72, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26101321

ABSTRACT

Considerable cross-talk exists between mechanisms controlling genome architecture and gene expression. AgR loci are excellent models for these processes because they are regulated at both conformational and transcriptional levels to facilitate their assembly by V(D)J recombination. Upon commitment to the double-negative stage of T cell development, Tcrb adopts a compact conformation that promotes long-range recombination between Vß gene segments (Trbvs) and their DßJß targets. Formation of a functional VßDßJß join signals for robust proliferation of double-negative thymocytes and their differentiation into double-positive (DP) cells, where Trbv recombination is squelched (allelic exclusion). DP differentiation also is accompanied by decontraction of Tcrb, which has been thought to separate the entire Trbv cluster from DßJß segments (spatial segregation-based model for allelic exclusion). However, DP cells also repress transcription of unrearranged Trbvs, which may contribute to allelic exclusion. We performed a more detailed study of developmental changes in Tcrb topology and found that only the most distal portion of the Trbv cluster separates from DßJß segments in DP thymocytes, leaving most Trbvs spatially available for rearrangement. Preferential dissociation of distal Trbvs is independent of robust proliferation or changes in transcription, chromatin, or architectural factors, which are coordinately regulated across the entire Trbv cluster. Segregation of distal Trbvs also occurs on alleles harboring a functional VßDßJß join, suggesting that this process is independent of rearrangement status and is DP intrinsic. Our finding that most Trbvs remain associated with DßJß targets in DP cells revises allelic exclusion models from their current conformation-dominant to a transcription-dominant formulation.


Subject(s)
Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/genetics , Genes, T-Cell Receptor beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Thymocytes/cytology , V(D)J Recombination/genetics , Animals , Base Sequence , Cell Differentiation/immunology , Cell Proliferation/genetics , Cyclin D3/genetics , High-Throughput Nucleotide Sequencing , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Conformation , Protein Structure, Secondary , Sequence Analysis, DNA
3.
Cell Cycle ; 14(3): 388-98, 2015.
Article in English | MEDLINE | ID: mdl-25659036

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is a cancer of immature T cells that exhibits heterogeneity of oncogenic lesions, providing an obstacle for development of more effective and less toxic therapies. Inherited deficiency of ATM, a regulator of the cellular DNA damage response, predisposes young humans and mice to T-ALLs with clonal chromosome translocations. While acquired ATM mutation or deletion occurs in pediatric T-ALLs, the role of somatic ATM alterations in T-ALL pathogenesis remains unknown. We demonstrate here that somatic Atm inactivation in haematopoietic cells starting as these cells differentiate in utero predisposes mice to T-ALL at similar young ages and harboring analogous translocations as germline Atm-deficient mice. However, some T-ALLs from haematopoietic cell specific deletion of Atm were of more mature thymocytes, revealing that the developmental timing and celluar origin of Atm inactivation influences the phenotype of ATM-deficient T-ALLs. Although it has been hypothesized that ATM suppresses cancer by preventing deletion and inactivation of TP53, we find that Atm inhibits T-ALL independent of Tp53 deletion. Finally, we demonstrate that the Cyclin D3 protein that drives immature T cell proliferation is essential for transformation of Atm-deficient thymocytes. Our study establishes a pre-clinical model for pediatric T-ALLs with acquired ATM inactivation and identifies the cell cycle machinery as a therapeutic target for this aggressive childhood T-ALL subtype.


Subject(s)
Cyclin D3/metabolism , Gene Silencing , Hematopoietic Stem Cells/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Disease Susceptibility , Gene Deletion , Genomic Instability , Heterozygote , Mice, Inbred C57BL , Mice, Transgenic , Thymocytes/metabolism , Tumor Suppressor Protein p53/metabolism
4.
J Exp Med ; 212(1): 107-20, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25512470

ABSTRACT

Gene regulation relies on dynamic changes in three-dimensional chromatin conformation, which are shaped by composite regulatory and architectural elements. However, mechanisms that govern such conformational switches within chromosomal domains remain unknown. We identify a novel mechanism by which cis-elements promote long-range interactions, inducing conformational changes critical for diversification of the TCRß antigen receptor locus (Tcrb). Association between distal Vß gene segments and the highly expressed DßJß clusters, termed the recombination center (RC), is independent of enhancer function and recruitment of V(D)J recombinase. Instead, we find that tissue-specific folding of Tcrb relies on two distinct architectural elements located upstream of the RC. The first, a CTCF-containing element, directly tethers distal portions of the Vß array to the RC. The second element is a chromatin barrier that protects the tether from hyperactive RC chromatin. When the second element is removed, active RC chromatin spreads upstream, forcing the tether to serve as a new barrier. Acquisition of barrier function by the CTCF element disrupts contacts between distal Vß gene segments and significantly alters Tcrb repertoires. Our findings reveal a separation of function for RC-flanking regions, in which anchors for long-range recombination must be cordoned off from hyperactive RC landscapes by chromatin barriers.


Subject(s)
Chromatin/genetics , Precursor Cells, B-Lymphoid/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Thymocytes/metabolism , Animals , CCCTC-Binding Factor , Cell Lineage/genetics , Cells, Cultured , Chromatin/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Histones/metabolism , In Situ Hybridization, Fluorescence/methods , Methylation , Mice, Inbred C57BL , Mice, Knockout , Precursor Cells, B-Lymphoid/cytology , Promoter Regions, Genetic/genetics , Protein Binding , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thymocytes/cytology , V(D)J Recombination/genetics , VDJ Recombinases/metabolism
5.
Cell Cycle ; 13(19): 3076-82, 2014.
Article in English | MEDLINE | ID: mdl-25486566

ABSTRACT

ATM is the master regulator of the cellular response to DNA double strand breaks (DSBs). Deficiency of ATM predisposes humans and mice to αß T lymphoid cancers with clonal translocations between the T cell receptor (TCR) α/δ locus and a 450 kb region of synteny on human chromosome 14 and mouse chromosome 12. While these translocations target and activate the TCL1 oncogene at 14q32 to cause T cell pro-lymphocytic leukemia (T-PLL), the TCRα/δ;14q32 translocations in ATM-deficient T cell acute lymphoblastic leukemia (T-ALL) have not been characterized and their role in cancer pathogenesis remains unknown. The corresponding lesion in Atm-deficient mouse T-ALLs is a chromosome t(12;14) translocation with Tcrδ genes fused to sequences on chromosome 12; although these translocations do not activate Tcl1, they delete the Bcl11b haploinsufficient tumor suppressor gene. To assess whether Tcrδ translocations that inactivate one copy of Bcl11b promote transformation of Atm-deficient cells, we analyzed Atm(-/-) mice with mono-allelic Bcl11b deletion initiating in thymocytes concomitant with Tcrδ recombination. Inactivation of one Bcl11b copy had no effect on the predisposition of Atm(-/-) mice to clonal T-ALLs. Yet, none of these T-ALLs had a clonal chromosome t(12;14) translocation that deleted Bcl11b indicating that Tcrδ translocations that inactivate a copy of Bcl11b promote transformation of Atm-deficient thymocytes. Our data demonstrate that antigen receptor locus translocations can cause cancer by deleting a tumor suppressor gene. We discuss the implications of these findings for the etiology and therapy of T-ALLs associated with ATM deficiency and TCRα/δ translocations targeting the 14q32 cytogenetic region.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Repressor Proteins/genetics , Translocation, Genetic , Tumor Suppressor Proteins/genetics , Alleles , Animals , Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Chromosomes , Flow Cytometry , Humans , Karyotyping , Metaphase , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism
6.
J Immunol ; 193(6): 2881-90, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25127855

ABSTRACT

Coordination of V rearrangements between loci on homologous chromosomes is critical for Ig and TCR allelic exclusion. The Ataxia Telangietasia mutated (ATM) protein kinase promotes DNA repair and activates checkpoints to suppress aberrant Ig and TCR rearrangements. In response to RAG cleavage of Igκ loci, ATM inhibits RAG expression and suppresses further Vκ-to-Jκ rearrangements to enforce Igκ allelic exclusion. Because V recombination between alleles is more strictly regulated for TCRß and IgH loci, we evaluated the ability of ATM to restrict biallelic expression and V-to-DJ recombination of TCRß and IgH genes. We detected greater frequencies of lymphocytes with biallelic expression or aberrant V-to-DJ rearrangement of TCRß or IgH loci in mice lacking ATM. A preassembled DJß complex that decreases the number of TCRß rearrangements needed for a productive TCRß gene further increased frequencies of ATM-deficient cells with biallelic TCRß expression. IgH and TCRß proteins drive proliferation of prolymphocytes through cyclin D3 (Ccnd3), which also inhibits VH transcription. We show that inactivation of Ccnd3 leads to increased frequencies of lymphocytes with biallelic expression of IgH or TCRß genes. We also show that Ccnd3 inactivation cooperates with ATM deficiency to increase the frequencies of cells with biallelic TCRß or IgH expression while decreasing the frequency of ATM-deficient lymphocytes with aberrant V-to-DJ recombination. Our data demonstrate that core components of the DNA damage response and cell cycle machinery cooperate to help enforce IgH and TCRß allelic exclusion and indicate that control of V-to-DJ rearrangements between alleles is important to maintain genomic stability.


Subject(s)
Cyclin D3/genetics , DNA Repair/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Proliferation , Cyclin D3/antagonists & inhibitors , DNA Damage , DNA Repair/immunology , Homeodomain Proteins/biosynthesis , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Heavy Chains/genetics , Immunoglobulin J-Chains/genetics , Immunoglobulin Variable Region/genetics , Immunoglobulin delta-Chains/genetics , Immunoglobulin kappa-Chains/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/immunology , VDJ Exons/genetics
7.
Cell Cycle ; 12(20): 3307-16, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24036547

ABSTRACT

TP53 protects cells from transformation by responding to stresses including aneuploidy and DNA double-strand breaks (DSBs). TP53 induces apoptosis of lymphocytes with persistent DSBs at antigen receptor loci and other genomic loci to prevent these lesions from generating oncogenic translocations. Despite this critical function of TP53, germline Tp53(-/-) mice succumb to immature T-cell (thymic) lymphomas that exhibit aneuploidy and lack clonal translocations. However, Tp53(-/-) mice occasionally develop B lineage lymphomas and Tp53 deletion in pro-B cells causes lymphomas with oncogenic immunoglobulin (Ig) locus translocations. In addition, human lymphoid cancers with somatic TP53 inactivation often harbor oncogenic IG or T-cell receptor (TCR) locus translocations. To determine whether somatic Tp53 inactivation unmasks translocations or alters the frequency of B lineage tumors in mice, we generated and analyzed mice with conditional Tp53 deletion initiating in hematopoietic stem cells (HSCs) or in lineage-committed thymocytes. Median tumor-free survival of each strain was similar to the lifespan of Tp53(-/-) mice. Mice with HSC deletion of Tp53 predominantly succumbed to thymic lymphomas with clonal translocations not involving Tcr loci; however, these mice occasionally developed mature B-cell lymphomas that harbored clonal Ig translocations. Deletion of Tp53 in thymocytes caused thymic lymphomas with aneuploidy and/or clonal translocations, including oncogenic Tcr locus translocations. Our data demonstrate that the developmental stage of Tp53 inactivation affects karyotypes of lymphoid malignancies in mice where somatic deletion of Tp53 initiating in thymocytes is sufficient to cause thymic lymphomas with oncogenic translocations.


Subject(s)
Gene Silencing , Genetic Predisposition to Disease , Hematopoietic Stem Cells/metabolism , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/pathology , Thymocytes/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Cell Lineage , Clone Cells , Gene Deletion , Hematopoietic Stem Cells/pathology , Humans , Mice , Thymocytes/pathology
8.
Cell Cycle ; 12(17): 2867-75, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23966158

ABSTRACT

The DNA damage response (DDR) can restrain the ability of oncogenes to cause genomic instability and drive malignant transformation. The gene encoding the histone H2AX DDR factor maps to 11q23, a region frequently altered in human cancers. Since H2ax functions as a haploinsufficient suppressor of B lineage lymphomas with c-Myc amplification and/or translocation, we determined the impact of H2ax expression on the ability of deregulated c-Myc expression to cause genomic instability and drive transformation of B cells. Neither H2ax deficiency nor haploinsufficiency affected the rate of mortality of Eµ-c-Myc mice from B lineage lymphomas with genomic deletions and amplifications. Yet H2ax functioned in a dosage-dependent manner to prevent unbalanced translocations in Eµ-c-Myc tumors, demonstrating that H2ax functions in a haploinsufficient manner to suppress allelic imbalances and limit molecular heterogeneity within and among Eµ-c-Myc lymphomas. Regardless of H2ax copy number, all Eµ-c-Myc tumors contained identical amplification of chromosome 19 sequences spanning 20 genes. Many of these genes encode proteins with tumor-promoting activities, including Cd274, which encodes the PD-L1 programmed death ligand that induces T cell apoptosis and enables cancer cells to escape immune surveillance. This amplicon was in non-malignant B and T cells and non-lymphoid cells, linked to the Eµ-c-Myc transgene, and associated with overexpression of PD-L1 on non-malignant B cells. Our data demonstrate that, in addition to deregulated c-Myc expression, non-malignant B lineage lymphocytes of Eµ-c-Myc transgenic mice may have constitutive amplification and increased expression of other tumor-promoting genes.


Subject(s)
Germ Cells/metabolism , Histones/metabolism , Lymphoma/genetics , Oncogenes/genetics , Proto-Oncogene Proteins c-myc/genetics , Translocation, Genetic , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Lineage/genetics , Chromosomes, Mammalian/metabolism , Clone Cells , Comparative Genomic Hybridization , Gene Amplification/genetics , Gene Deletion , Haploinsufficiency/genetics , Humans , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Suppression, Genetic , Transgenes/genetics
9.
J Exp Med ; 210(2): 233-9, 2013 Feb 11.
Article in English | MEDLINE | ID: mdl-23382544

ABSTRACT

Allelic exclusion is enforced through the ability of antigen receptor chains expressed from one allele to signal feedback inhibition of V-to-(D)J recombination on the other allele. To achieve allelic exclusion by such means, only one allele can initiate V-to-(D)J recombination within the time required to signal feedback inhibition. DNA double-strand breaks (DSBs) induced by the RAG endonuclease during V(D)J recombination activate the Ataxia Telangiectasia mutated (ATM) and DNA-dependent protein kinase (DNA-PK) kinases. We demonstrate that ATM enforces Igκ allelic exclusion, and that RAG DSBs induced during Igκ recombination in primary pre-B cells signal through ATM, but not DNA-PK, to suppress initiation of additional Igκ rearrangements. ATM promotes high-density histone H2AX phosphorylation to create binding sites for MDC1, which functions with H2AX to amplify a subset of ATM-dependent signals. However, neither H2AX nor MDC1 is required for ATM to enforce Igκ allelic exclusion and suppress Igκ rearrangements. Upon activation in response to RAG Igκ cleavage, ATM signals down-regulation of Gadd45α with concomitant repression of the Gadd45α targets Rag1 and Rag2. Our data indicate that ATM kinases activated by RAG DSBs during Igκ recombination transduce transient H2AX/MDC1-independent signals that suppress initiation of further Igκ rearrangements to control Igκ allelic exclusion.


Subject(s)
Cell Cycle Proteins/immunology , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Gene Rearrangement, B-Lymphocyte, Light Chain , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/immunology , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing , Alleles , Animals , Ataxia Telangiectasia Mutated Proteins , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Base Sequence , Cell Cycle Proteins/genetics , DNA Breaks, Double-Stranded , DNA-Binding Proteins/genetics , Histones/deficiency , Histones/genetics , Histones/metabolism , Homeodomain Proteins/metabolism , Immunoglobulin kappa-Chains/genetics , Immunoglobulin kappa-Chains/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, 129 Strain , Mice, Knockout , Models, Biological , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Tumor Suppressor Proteins/genetics
10.
Cancer Immunol Res ; 1(6): 426-37, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24778134

ABSTRACT

Drosophila melanogaster discs large (dlg) is an essential tumor suppressor gene (TSG) controlling epithelial cell growth and polarity of the fly imaginal discs in pupal development. A mammalian ortholog, Dlg1, is involved in embryonic urogenital morphogenesis, postsynaptic densities in neurons, and immune synapses in lymphocytes. However, a potential role for Dlg1 as a mammalian TSG is unknown. Here, we present evidence that loss of Dlg1 confers strong predisposition to the development of malignancies in a murine model of pediatric B-cell acute lymphoblastic leukemia (B-ALL). Using mice with conditionally deleted Dlg1 alleles, we identify a novel "pre-leukemic" stage of developmentally arrested early B-lineage cells marked by preeminent c-Myc expression. Mechanistically, we show that in B-lineage progenitors Dlg1 interacts with and stabilizes the PTEN protein, regulating its half-life and steady-state abundance. The loss of Dlg1 does not affect the level of PTEN mRNAs but results in a dramatic decrease in PTEN protein, leading to excessive phosphoinositide 3-kinase signaling and proliferation. Our data suggest a novel model of tumor suppression by a PDZ domain-containing polarity gene in hematopoietic cancers.


Subject(s)
Genes, Tumor Suppressor/physiology , Nerve Tissue Proteins/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Discs Large Homolog 1 Protein , Disease Models, Animal , Genetic Predisposition to Disease , Mice, Knockout , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/metabolism , PTEN Phosphohydrolase/biosynthesis , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Receptors, Interleukin-7/physiology , SAP90-PSD95 Associated Proteins , Signal Transduction/genetics , Tumor Suppressor Protein p53/deficiency
11.
J Immunol ; 189(3): 1372-9, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22730535

ABSTRACT

The ataxia telangiectasia mutated (ATM) kinase and H2AX histone tumor suppressor proteins are each critical for maintenance of cellular genomic stability and suppression of lymphomas harboring clonal translocations. ATM is the predominant kinase that phosphorylates H2AX in chromatin around DNA double-strand breaks, including along lymphocyte Ag receptor loci cleaved during V(D)J recombination. However, combined germline inactivation of Atm and H2ax in mice causes early embryonic lethality associated with substantial cellular genomic instability, indicating that ATM and H2AX exhibit nonredundant functions in embryonic cells. To evaluate potential nonredundant roles of ATM and H2AX in somatic cells, we generated and analyzed Atm-deficient mice with conditional deletion of H2ax in αß T-lineage lymphocytes. Combined Atm/H2ax inactivation starting in early-stage CD4(-)/CD8(-) thymocytes resulted in lower numbers of later-stage CD4(+)/CD8(+) thymocytes, but led to no discernible V(D)J recombination defect in G1 phase cells beyond that observed in Atm-deficient cells. H2ax deletion in Atm-deficient thymocytes also did not affect the incidence or mortality of mice from thymic lymphomas with clonal chromosome 14 (TCRα/δ) translocations. Yet, in vitro-stimulated Atm/H2ax-deficient splenic αß T cells exhibited a higher frequency of genomic instability, including radial chromosome translocations and TCRß translocations, compared with cells lacking Atm or H2ax. Collectively, our data demonstrate that both redundant and nonredundant functions of ATM and H2AX are required for normal recombination of TCR loci, proliferative expansion of developing thymocytes, and maintenance of genomic stability in cycling αß T-lineage cells.


Subject(s)
Ataxia Telangiectasia/immunology , Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , Histones/physiology , Protein Serine-Threonine Kinases/physiology , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocyte Subsets/immunology , Tumor Suppressor Proteins/physiology , Animals , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/pathology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle/genetics , Cell Cycle/immunology , Cell Cycle Proteins/deficiency , Cell Lineage/genetics , Cell Lineage/immunology , Cell Proliferation , DNA-Binding Proteins/deficiency , Genetic Loci/immunology , Genomic Instability/immunology , Histones/deficiency , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Serine-Threonine Kinases/deficiency , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Recombination, Genetic/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Tumor Suppressor Proteins/deficiency
12.
Blood ; 117(1): 175-85, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-20947684

ABSTRACT

H2AX and Artemis each cooperate with p53 to suppress lymphoma. Germline H2ax(-/-)p53(-/-) mice die of T-cell receptor-ß(-) (TCR-ß(-)) thymic lymphomas with translocations and other lesions characteristic of human T-cell acute lymphoblastic leukemia. Here, we demonstrate that mice with inactivation of H2ax and p53 in thymocytes die at later ages to TCR-ß(-) or TCR-ß(+) thymic lymphomas containing a similar pattern of translocations as H2ax(-/-)p53(-/-) tumors. Germline Artemis(-/-) p53(-/-) mice die of lymphomas with antigen receptor locus translocations, whereas Artemis(-/-)H2ax(-/-)p53(-/-) mice die at earlier ages from multiple malignancies. We show here that Artemis(-/-) mice with p53 deletion in thymocytes die of TCR-ß(-) tumors containing Tcrα/δ translocations, other clonal translocations, or aneuploidy, as well as Notch1 mutations. Strikingly, Artemis(-/-) mice with H2ax and p53 deletion in thymocytes exhibited a lower rate of mortality from TCR-ß(-) tumors, which harbored significantly elevated levels of genomic instability. Our data reveal that the cellular origin of H2ax and p53 loss impacts the rate of mortality from and developmental stage of thymic lymphomas, and suggest that conditional deletion of tumor suppressor genes may provide more physiologic models for human lymphoid malignancies than germline inactivation.


Subject(s)
Histones/physiology , Lymphoma/pathology , Sequence Deletion , Thymus Neoplasms/pathology , Tumor Suppressor Protein p53/physiology , Animals , Blotting, Southern , Blotting, Western , Endonucleases , Flow Cytometry , Genomic Instability , Humans , In Situ Hybridization, Fluorescence , Lymphoma/etiology , Lymphoma/metabolism , Mice , Mice, Knockout , Nuclear Proteins/physiology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Thymus Neoplasms/etiology , Thymus Neoplasms/metabolism , Translocation, Genetic
13.
J Immunol ; 185(6): 3564-73, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20709953

ABSTRACT

The genomic organization of TCRbeta loci enables Vbeta-to-DJbeta2 rearrangements on alleles with assembled VbetaDJbetaCbeta1 genes, which could have deleterious physiologic consequences. To determine whether such Vbeta rearrangements occur and, if so, how they might be regulated, we analyzed mice with TCRbeta alleles containing preassembled functional VbetaDJbetaCbeta1 genes. Vbeta10 segments were transcribed, rearranged, and expressed in thymocytes when located immediately upstream of a Vbeta1DJbetaCbeta1 gene, but not on alleles with a Vbeta14DJbetaCbeta1 gene. Germline Vbeta10 transcription was silenced in mature alphabeta T cells. This allele-dependent and developmental stage-specific silencing of Vbeta10 correlated with increased CpG methylation and decreased histone acetylation over the Vbeta10 promoter and coding region. Transcription, rearrangement, and expression of the Vbeta4 and Vbeta16 segments located upstream of Vbeta10 were silenced on alleles containing either VbetaDJbetaCbeta1 gene; sequences within Vbeta4, Vbeta16, and the Vbeta4/Vbeta16-Vbeta10 intergenic region exhibited constitutive high CpG methylation and low histone acetylation. Collectively, our data indicate that the position of Vbeta segments relative to assembled VbetaDJbetaCbeta1 genes influences their rearrangement and suggest that DNA sequences between Vbeta segments may form boundaries between active and inactive Vbeta chromatin domains upstream of VbetaDJbetaCbeta genes.


Subject(s)
Alleles , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/immunology , Gene Silencing/immunology , Genes, T-Cell Receptor beta/physiology , Germ-Line Mutation/immunology , Peptide Fragments/genetics , Receptors, Antigen, T-Cell, alpha-beta/antagonists & inhibitors , Receptors, Antigen, T-Cell, alpha-beta/genetics , Animals , Chromatin/chemistry , Chromatin/genetics , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Hybridomas , Immunoglobulin Variable Region/biosynthesis , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , Immunosuppression Therapy/methods , Mice , Mice, Knockout , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Transcription, Genetic/immunology
14.
J Immunol ; 185(2): 1055-62, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20562258

ABSTRACT

Feedback inhibition of V(D)J recombination enforces Ag receptor allelic exclusion in mammalian lymphocytes. Yet, in-frame VbetaDJbeta exons can assemble on both alleles in human and mouse alphabeta T lineage cells. To elucidate mechanisms that enforce TCRbeta allelic exclusion in such cells, we analyzed Vbeta expression and rearrangement in mice containing a functional Vbeta14DJbeta1.5Cbeta1 gene (Vbeta14(NT)) and/or Vbeta8.2DJbeta1.1Cbeta1 transgene (Vbeta8(Tg)). The majority of Vbeta14(NT) and Vbeta8(Tg) alphabeta T lineage cells expressed only Vbeta14(+) or Vbeta8(+) TCRbeta-chains, respectively, and lacked Vbeta rearrangements on wild-type TCRbeta loci. However, endogenous Vbeta rearrangements and alphabeta T lineage cells expressing endogenous Vbetas from wild-type alleles alone or with the prerearranged Vbeta in cell surface TCRbeta-chains were observed in Vbeta14(NT) and Vbeta8(Tg) mice. Although nearly all Vbeta8(Tg):Vbeta14(NT) thymocytes and splenic alphabeta T cells expressed Vbeta8(+) TCRbeta-chains, only half of these lymphocytes expressed Vbeta14(+) TCRbeta-chains, even though similar steady-state levels of Vbeta14(NT) mRNA were expressed in Vbeta8(+)Vbeta14(+) and Vbeta8(+)Vbeta14(-) populations. Our data demonstrated that posttranscriptional silencing of functionally assembled endogenous VbetaDJbetaCbeta genes can enforce TCRbeta allelic exclusion and reveal another mechanism that contributes to the development of lymphocytes with monospecific Ag receptors.


Subject(s)
RNA Interference , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/metabolism , Alleles , Animals , Flow Cytometry , Gene Rearrangement, T-Lymphocyte , Hybridomas/metabolism , Mice , Mice, Inbred Strains , Mice, Knockout , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/cytology
15.
J Immunol ; 184(3): 1369-78, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20042591

ABSTRACT

Ag receptor allelic exclusion is thought to occur through monoallelic initiation and subsequent feedback inhibition of recombinational accessibility. However, our previous analysis of mice containing a V(D)J recombination reporter inserted into Vbeta14 (Vbeta14(Rep)) indicated that Vbeta14 chromatin accessibility is biallelic. To determine whether Vbeta14 recombinational accessibility is subject to feedback inhibition, we analyzed TCRbeta rearrangements in Vbeta14(Rep) mice containing a preassembled in-frame transgenic Vbeta8.2Dbeta1Jbeta1.1 or an endogenous Vbeta14Dbeta1Jbeta1.4 rearrangement on the homologous chromosome. Expression of either preassembled VbetaDJbetaC beta-chain accelerated thymocyte development because of enhanced cellular selection, demonstrating that the rate-limiting step in early alphabeta T cell development is the assembly of an in-frame VbetaDJbeta rearrangement. Expression of these preassembled VbetaDJbeta rearrangements inhibited endogenous Vbeta14-to-DJbeta rearrangements as expected. However, in contrast to results predicted by the accepted model of TCRbeta feedback inhibition, we found that expression of these preassembled TCR beta-chains did not downregulate recombinational accessibility of Vbeta14 chromatin. Our findings suggest that TCRbeta-mediated feedback inhibition of Vbeta14 rearrangements depends on inherent properties of Vbeta14, Dbeta, and Jbeta recombination signal sequences.


Subject(s)
Antibody Diversity , Chromatin/physiology , Feedback, Physiological/physiology , Gene Rearrangement, T-Lymphocyte/immunology , Immunoglobulin Joining Region , Receptors, Antigen, T-Cell, alpha-beta/antagonists & inhibitors , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Signal Transduction/immunology , Animals , Antibody Diversity/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Gene Expression Regulation, Developmental/immunology , Genes, Reporter/immunology , Germ-Line Mutation/immunology , Immunoglobulin Joining Region/genetics , Immunoglobulin Variable Region/genetics , Loss of Heterozygosity/immunology , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/genetics , Signal Transduction/genetics , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
16.
J Exp Med ; 206(12): 2625-39, 2009 Nov 23.
Article in English | MEDLINE | ID: mdl-19887394

ABSTRACT

The H2AX core histone variant is phosphorylated in chromatin around DNA double strand breaks (DSBs) and functions through unknown mechanisms to suppress antigen receptor locus translocations during V(D)J recombination. Formation of chromosomal coding joins and suppression of translocations involves the ataxia telangiectasia mutated and DNA-dependent protein kinase catalytic subunit serine/threonine kinases, each of which phosphorylates H2AX along cleaved antigen receptor loci. Using Abelson transformed pre-B cell lines, we find that H2AX is not required for coding join formation within chromosomal V(D)J recombination substrates. Yet we show that H2AX is phosphorylated along cleaved Igkappa DNA strands and prevents their separation in G1 phase cells and their progression into chromosome breaks and translocations after cellular proliferation. We also show that H2AX prevents chromosome breaks emanating from unrepaired RAG endonuclease-generated TCR-alpha/delta locus coding ends in primary thymocytes. Our data indicate that histone H2AX suppresses translocations during V(D)J recombination by creating chromatin modifications that stabilize disrupted antigen receptor locus DNA strands to prevent their irreversible dissociation. We propose that such H2AX-dependent mechanisms could function at additional chromosomal locations to facilitate the joining of DNA ends generated by other types of DSBs.


Subject(s)
Chromatin/metabolism , Chromosome Breakage , DNA Breaks, Double-Stranded , Histones/metabolism , Recombination, Genetic/physiology , VDJ Exons/physiology , Animals , Ataxia Telangiectasia Mutated Proteins , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/immunology , Cell Cycle Proteins/metabolism , Cell Line, Transformed , Chromatin/genetics , Chromatin/immunology , G1 Phase/physiology , Histones/genetics , Histones/immunology , Mice , Mice, Knockout , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Quantitative Trait Loci/physiology , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism
17.
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
18.
J Immunol ; 182(9): 5586-95, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19380806

ABSTRACT

TCRbeta chain repertoire of peripheral alphabeta T cells is generated through the stepwise assembly and subsequent selection of TCRbeta V region exons during thymocyte development. To evaluate the influence of a two-step recombination process on Vbeta rearrangement and selection, we generated mice with a preassembled Dbeta1Jbeta1.1 complex on the Jbeta1(omega) allele, an endogenous TCRbeta allele that lacks the Dbeta2-Jbeta2 cluster, creating the Jbeta1(DJbeta) allele. As compared with Jbeta1(omega/omega) mice, both Jbeta1(DJbeta/omega) and Jbeta1(DJbeta/DJbeta) mice exhibited grossly normal thymocyte development and TCRbeta allelic exclusion. In addition, Vbeta rearrangements on Jbeta1(DJbeta) and Jbeta1(omega) alleles were similarly regulated by TCRbeta-mediated feedback regulation. However, in-frame VbetaDJbeta rearrangements were present at a higher level on the Jbeta1(DJbeta) alleles of Jbeta1(DJbeta/omega) alphabeta T cell hybridomas, as compared with on the Jbeta1(omega) alleles. This bias was most likely due to both an increased frequency of Vbeta-to-DJbeta rearrangements on Jbeta1(DJbeta) alleles and a preferential selection of cells with in-frame VbetaDJbeta exons assembled on Jbeta1(DJbeta) alleles during the development of Jbeta1(DJbeta/omega) alphabeta T cells. Consistent with the differential selection of in-frame VbetaDJbeta rearrangements on Jbeta1(DJbeta) alleles, the Vbeta repertoire of alphabeta T cells was significantly altered during alphabeta TCR selection in Jbeta1(DJbeta/omega) and Jbeta1(DJbeta/DJbeta) mice, as compared with in Jbeta1(omega/omega) mice. Our data indicate that the diversity of DJbeta complexes assembled during thymocyte development influences TCRbeta chain selection and peripheral Vbeta repertoire.


Subject(s)
Antibody Diversity/genetics , Gene Rearrangement, T-Lymphocyte/immunology , Immunoglobulin Joining Region/genetics , Immunoglobulin Variable Region/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Alleles , Animals , Antibody Diversity/immunology , Base Sequence , Cell Line, Tumor , Cells, Cultured , Gene Targeting , Immunoglobulin Joining Region/biosynthesis , Immunoglobulin Variable Region/biosynthesis , Mice , Mice, Transgenic , Molecular Sequence Data , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Recombination, Genetic , T-Lymphocyte Subsets/cytology
19.
Eur J Immunol ; 38(9): 2564-72, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18792409

ABSTRACT

T-cell receptor (TCR) beta variable region exons are assembled from numerous gene segments in a highly ordered and regulated manner. To elucidate mechanisms and identify cis-acting elements that control Vbeta rearrangement, we generated an endogenous TCR-beta allele with only the Vbeta2, Vbeta4, and Vbeta14 segments. We found that alphabeta T lineage cells containing this Vbeta(2-4-14) allele and a wild-type TCR-beta allele developed normally, but exhibited a significant increase in Vbeta2(+) and Vbeta14(+) cells. To quantify Vbeta rearrangements on the Vbeta(2-4-14) allele, we generated alphabeta T-cell hybridomas and analyzed TCR-beta rearrangements. Despite the deletion of almost all Vbeta segments and 234 kb of Vbeta cluster sequences, the Vbeta(2-4-14) allele exhibited only a slight decrease in Vbeta rearrangement as compared with the wild-type TCR-beta allele. Thus, cis-acting control elements essential for directing Vbeta rearrangement across large chromosomal distances are not located within the Vbeta cluster. We also found a significant increase in the frequency of Vbeta rearrangements involving Vbeta2 and Vbeta14, but not Vbeta4, on the Vbeta(2-4-14) allele. Collectively, our data suggest that Vbeta cluster sequences reduce the frequency of Vbeta2 and Vbeta14 rearrangements by competing with the productive coupling of accessible Vbeta2 and Vbeta14 segments with DJbeta1 complexes.


Subject(s)
Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Genes, T-Cell Receptor beta , Receptors, Antigen, T-Cell, alpha-beta/genetics , Alleles , Animals , Mice , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...