Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 117(10): 5453-5462, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32098847

ABSTRACT

Developing lymphocytes diversify their antigen receptor (AgR) loci by variable (diversity) joining (V[D]J) recombination. Here, using the micrococcal nuclease (MNase)-based chromatin accessibility (MACC) assay with low-cell count input, we profile both small-scale (kilobase) and large-scale (megabase) changes in chromatin accessibility and nucleosome occupancy in primary cells during lymphoid development, tracking the changes as different AgR loci become primed for recombination. The three distinct chromatin structures identified in this work define unique features of immunoglobulin H (IgH), Igκ, and T cell receptor-α (TCRα) loci during B lymphopoiesis. In particular, we find locus-specific temporal changes in accessibility both across megabase-long AgR loci and locally at the recombination signal sequences (RSSs). These changes seem to be regulated independently and can occur prior to lineage commitment. Large-scale changes in chromatin accessibility occur without significant change in nucleosome density and represent key features of AgR loci not previously described. We further identify local dynamic repositioning of individual RSS-associated nucleosomes at IgH and Igκ loci while they become primed for recombination during B cell commitment. These changes in chromatin at AgR loci are regulated in a locus-, lineage-, and stage-specific manner during B lymphopoiesis, serving either to facilitate or to impose a barrier to V(D)J recombination. We suggest that local and global changes in chromatin openness in concert with nucleosome occupancy and placement of histone modifications facilitate the temporal order of AgR recombination. Our data have implications for the organizing principles that govern assembly of these large loci as well as for mechanisms that might contribute to aberrant V(D)J recombination and the development of lymphoid tumors.


Subject(s)
B-Lymphocytes/physiology , Chromatin/metabolism , Gene Rearrangement, B-Lymphocyte , Lymphopoiesis/genetics , Receptors, Antigen/genetics , V(D)J Recombination , Animals , Chromatin/chemistry , Genetic Loci , Genetic Testing , Immunoglobulin Heavy Chains/genetics , Immunoglobulin kappa-Chains/genetics , Lymphoma/genetics , Mice , Mice, Inbred C57BL , Micrococcal Nuclease , Nucleosomes , Receptors, Antigen, T-Cell, alpha-beta/genetics
2.
J Biol Chem ; 294(37): 13580-13592, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31285261

ABSTRACT

Antigen receptor assembly in lymphocytes involves stringently-regulated coordination of specific DNA rearrangement events across several large chromosomal domains. Previous studies indicate that transcription factors such as paired box 5 (PAX5), Yin Yang 1 (YY1), and CCCTC-binding factor (CTCF) play a role in regulating the accessibility of the antigen receptor loci to the V(D)J recombinase, which is required for these rearrangements. To gain clues about the role of CTCF binding at the murine immunoglobulin heavy chain (IgH) locus, we utilized a computational approach that identified 144 putative CTCF-binding sites within this locus. We found that these CTCF sites share a consensus motif distinct from other CTCF sites in the mouse genome. Additionally, we could divide these CTCF sites into three categories: intergenic sites remote from any coding element, upstream sites present within 8 kb of the VH-leader exon, and recombination signal sequence (RSS)-associated sites characteristically located at a fixed distance (∼18 bp) downstream of the RSS. We noted that the intergenic and upstream sites are located in the distal portion of the VH locus, whereas the RSS-associated sites are located in the DH-proximal region. Computational analysis indicated that the prevalence of CTCF-binding sites at the IgH locus is evolutionarily conserved. In all species analyzed, these sites exhibit a striking strand-orientation bias, with >98% of the murine sites being present in one orientation with respect to VH gene transcription. Electrophoretic mobility shift and enhancer-blocking assays and ChIP-chip analysis confirmed CTCF binding to these sites both in vitro and in vivo.


Subject(s)
CCCTC-Binding Factor/metabolism , Immunoglobulin Heavy Chains/genetics , Adaptive Immunity/genetics , Animals , Binding Sites , CCCTC-Binding Factor/genetics , Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/genetics , Gene Rearrangement , Humans , Immunoglobulin Heavy Chains/metabolism , Immunoglobulin Variable Region , K562 Cells , Mice , Mice, Knockout , NIH 3T3 Cells , Nucleotide Motifs , Regulatory Sequences, Nucleic Acid , Repressor Proteins/metabolism
3.
Curr Protoc Mol Biol ; 127(1): e91, 2019 06.
Article in English | MEDLINE | ID: mdl-31237423

ABSTRACT

An understanding of the dynamic structural properties of chromatin requires techniques that allow the profiling of regions of both open and closed chromatin as well as the assessment of nucleosome occupancy. The recently developed MNase accessibility (MACC) technique allows for the simultaneous measurement of chromatin opening and compaction, as well as nucleosome occupancy, on a genome-wide scale in a single assay. This article presents a low-input MACC procedure that considerably extends the utility of the original MACC assay. Low-input MACC generates high-quality data using very low cell numbers (as few as 50 cells per titration point), making it ideal for samples obtained after fluorescence-activated cell sorting or dissection, or in clinical settings. Moreover, low-input MACC has significantly improved several steps of the initial method, offering a more rapid and robust methodology. © 2019 by John Wiley & Sons, Inc.


Subject(s)
Chromatin/chemistry , Eukaryotic Cells/metabolism , High-Throughput Nucleotide Sequencing/methods , Micrococcal Nuclease/metabolism , Nucleosomes , Chromatin Assembly and Disassembly , Nucleosomes/genetics , Nucleosomes/metabolism
4.
Cancer Discov ; 7(11): 1336-1353, 2017 11.
Article in English | MEDLINE | ID: mdl-28974511

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy of thymocytes. Using a transgenic screen in zebrafish, thymocyte selection-associated high mobility group box protein (TOX) was uncovered as a collaborating oncogenic driver that accelerated T-ALL onset by expanding the initiating pool of transformed clones and elevating genomic instability. TOX is highly expressed in a majority of human T-ALL and is required for proliferation and continued xenograft growth in mice. Using a wide array of functional analyses, we uncovered that TOX binds directly to KU70/80 and suppresses recruitment of this complex to DNA breaks to inhibit nonhomologous end joining (NHEJ) repair. Impaired NHEJ is well known to cause genomic instability, including development of T-cell malignancies in KU70- and KU80-deficient mice. Collectively, our work has uncovered important roles for TOX in regulating NHEJ by elevating genomic instability during leukemia initiation and sustaining leukemic cell proliferation following transformation.Significance: TOX is an HMG box-containing protein that has important roles in T-ALL initiation and maintenance. TOX inhibits the recruitment of KU70/KU80 to DNA breaks, thereby inhibiting NHEJ repair. Thus, TOX is likely a dominant oncogenic driver in a large fraction of human T-ALL and enhances genomic instability. Cancer Discov; 7(11); 1336-53. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 1201.


Subject(s)
DNA End-Joining Repair/genetics , Genomic Instability/genetics , HMGB Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcription Factors/genetics , Animals , Animals, Genetically Modified , Cell Proliferation/genetics , Humans , Ku Autoantigen/genetics , Mice , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , T-Lymphocytes/pathology , Xenograft Model Antitumor Assays , Zebrafish/genetics
5.
Proc Natl Acad Sci U S A ; 113(42): E6427-E6436, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27698124

ABSTRACT

We show that the physical distribution of nucleosomes at antigen receptor loci is subject to regulated cell type-specific and lineage-specific positioning and correlates with the accessibility of these gene segments to recombination. At the Ig heavy chain locus (IgH), a nucleosome in pro-B cells is generally positioned over each IgH variable (VH) coding segment, directly adjacent to the recombination signal sequence (RSS), placing the RSS in a position accessible to the recombination activating gene (RAG) recombinase. These changes result in establishment of a specific chromatin organization at the RSS that facilitates accessibility of the genomic DNA for the RAG recombinase. In contrast, in mouse embryonic fibroblasts the coding segment is depleted of nucleosomes, which instead cover the RSS, thereby rendering it inaccessible. Pro-T cells exhibit a pattern intermediate between pro-B cells and mouse embryonic fibroblasts. We also find large-scale variations of nucleosome density over hundreds of kilobases, delineating chromosomal domains within IgH, in a cell type-dependent manner. These findings suggest that developmentally regulated changes in nucleosome location and occupancy, in addition to the known chromatin modifications, play a fundamental role in regulating V(D)J recombination. Nucleosome positioning-which has previously been observed to vary locally at individual enhancers and promoters-may be a more general mechanism by which cells can regulate the accessibility of the genome during development, at scales ranging from several hundred base pairs to many kilobases.


Subject(s)
Nucleosomes/metabolism , V(D)J Recombination , Animals , Cell Line , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , Chromosome Mapping , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Epigenomics , Gene Knockout Techniques , Genetic Loci , High-Throughput Nucleotide Sequencing , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Mice, Knockout , Organ Specificity , Precursor Cells, B-Lymphoid/metabolism , Protein Binding , Receptors, Antigen, T-Cell, alpha-beta/genetics
6.
PLoS One ; 10(4): e0121489, 2015.
Article in English | MEDLINE | ID: mdl-25849362

ABSTRACT

Omenn syndrome is a primary immunodeficiency disorder, featuring susceptibility to infections and autoreactive T cells and resulting from defective genomic rearrangement of genes for the T cell and B cell receptors. The most frequent etiologies are hypomorphic mutations in "non-core" regions of the Rag1 or Rag2 genes, the protein products of which are critical members of the cellular apparatus for V(D)J recombination. In this report, we describe an infant with Omenn syndrome with a previously unreported termination mutation (p.R142*) in Rag1 on one allele and a partially characterized substitution mutation (p.V779M) in a "core" region of the other Rag1 allele. Using a cellular recombination assay, we found that while the p.R142* mutation completely abolished V(D)J recombination activity, the p.V779M mutation conferred a severe, but not total, loss of V(D)J recombination activity. The recombination defect of the V779 mutant was not due to overall misfolding of Rag1, however, as this mutant supported wild-type levels of V(D)J cleavage. These findings provide insight into the role of this poorly understood region of Rag1 and support the role of Rag1 in a post-cleavage stage of recombination.


Subject(s)
Alleles , Heterozygote , Homeodomain Proteins/genetics , Mutation, Missense , Severe Combined Immunodeficiency/genetics , Amino Acid Substitution , Child , Child, Preschool , Homeodomain Proteins/metabolism , Humans , Infant , Infant, Newborn , Male , Proteolysis , Recombination, Genetic , Severe Combined Immunodeficiency/metabolism
7.
Cell Rep ; 1(2): 83-90, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-22720264

ABSTRACT

The preferential in vitro interaction of the PHD finger of RAG2, a subunit of the V(D)J recombinase, with histone H3 tails simultaneously trimethylated at lysine 4 and symmetrically dimethylated at arginine 2 (H3R2me2sK4me3) predicted the existence of the previously unknown histone modification H3R2me2s. Here, we report the in vivo identification of H3R2me2s . Consistent with the binding specificity of the RAG2 PHD finger, high levels of H3R2me2sK4me3 are found at antigen receptor gene segments ready for rearrangement. However, this double modification is much more general; it is conserved throughout eukaryotic evolution. In mouse, H3R2me2s is tightly correlated with H3K4me3 at active promoters throughout the genome. Mutational analysis in S. cerevisiae reveals that deposition of H3R2me2s requires the same Set1 complex that deposits H3K4me3. Our work suggests that H3R2me2sK4me3, not simply H3K4me3 alone, is the mark of active promoters and that factors that recognize H3K4me3 will have their binding modulated by their preference for H3R2me2s.


Subject(s)
Arginine/metabolism , Eukaryota/genetics , Eukaryota/metabolism , Genome/genetics , Histones/metabolism , Lysine/metabolism , Animals , Conserved Sequence/genetics , Evolution, Molecular , Genetic Loci/genetics , Histone-Lysine N-Methyltransferase/metabolism , Methylation , Mice , RNA, Small Interfering/metabolism , Receptors, Antigen/immunology , Recombination, Genetic/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
8.
Mol Immunol ; 52(1): 1-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22580346

ABSTRACT

Antibody switching involves class switch recombination (CSR) events between switch (S) regions located upstream of heavy chain constant (C) genes. Mechanisms targeting CSR to S-regions are not clear. Deletion of Sµ tandem repeat (SµTR) sequences causes CSR to shift into downstream regions that do not undergo CSR in WT B-cells, including the Cµ-region. We now find that, in SµTR(-/-) B cells, Sµ chromatin histone modification patterns also shift downstream relative to WT and coincide with SµTR(-/-) CSR locations. Our results suggest that histone H3 acetylation and methylation are involved in accessibility of switch regions and that these modifications are not dependent on the underlying sequence, but may be controlled by the location of upstream promoter or regulatory elements. Our studies also show RNA polymerase II (RNAPII) loading increases in the Eµ/Iµ region in stimulated B cells; these increases are independent of SµTR sequences. Longer Sµ deletions have been reported to eliminate increases in RNAPII density, therefore we suggest that sequences between Iµ and Sµ (possibly the Iµ splicing region as well as G-tracts that are involved in stable RNA:DNA complex formation during transcription) might control the RNAPII density increases.


Subject(s)
Immunoglobulin Class Switching/genetics , Immunoglobulin mu-Chains/genetics , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Base Sequence , Cells, Cultured , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/immunology , DNA Breaks , Gene Knockout Techniques , Histones/metabolism , Mice , Mice, Inbred C57BL , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Tandem Repeat Sequences
9.
Adv Exp Med Biol ; 650: 16-31, 2009.
Article in English | MEDLINE | ID: mdl-19731798

ABSTRACT

V(D)J recombination is initiated by the lymphoid specific proteins RAG1 and RAG2, which together constitute the V(D)J recombinase. However, the RAG 1/2 complex can also act as a transposase, inserting the broken DNA molecules generated during V(D)J recombination into an unrelated piece of DNA. This process, termed RAG transposition, can potentially cause insertional mutagenesis, chromosomal translocations and genomic instability. This review focuses on the mechanism and regulation of RAG transposition. We first provide a brief overview of the biochemistry of V(D)J recombination. We then discuss the discovery of RAG transposition and present an overview of the RAG transposition pathway. Using this pathway as a framework, we discuss the factors and forces that regulate RAG transposition.


Subject(s)
DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Mutagenesis, Insertional , Recombination, Genetic , Translocation, Genetic , Animals , DNA-Binding Proteins/genetics , Gene Rearrangement, B-Lymphocyte , Gene Rearrangement, T-Lymphocyte , Genes, RAG-1 , Humans , VDJ Recombinases/metabolism
10.
Nat Immunol ; 10(8): 817-21, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19621044

ABSTRACT

During B cell and T cell development, the lymphoid-specific proteins RAG-1 and RAG-2 act together to initiate the assembly of antigen receptor genes through a series of site-specific somatic DNA rearrangements that are collectively called variable-diversity-joining (V(D)J) recombination. In the past 20 years, a great deal has been learned about the enzymatic activities of the RAG-1-RAG-2 complex. Recent studies have identified several new and exciting regulatory functions of the RAG-1-RAG-2 complex. Here we discuss some of these functions and suggest that the RAG-1-RAG-2 complex nucleates a specialized subnuclear compartment that we call the 'V(D)J recombination factory'.


Subject(s)
DNA-Binding Proteins/immunology , Gene Rearrangement/immunology , Homeodomain Proteins/immunology , Models, Biological , Nuclear Proteins/immunology , Recombination, Genetic , VDJ Recombinases/immunology , Animals , B-Lymphocytes/immunology , Chromatin/metabolism , DNA Damage/immunology , DNA Repair/immunology , Histones/metabolism , Humans , Protein Binding
12.
Proc Natl Acad Sci U S A ; 104(48): 18993-8, 2007 Nov 27.
Article in English | MEDLINE | ID: mdl-18025461

ABSTRACT

Recombination activating gene (RAG) 1 and RAG2 together catalyze V(D)J gene rearrangement in lymphocytes as the first step in the assembly and maturation of antigen receptors. RAG2 contains a plant homeodomain (PHD) near its C terminus (RAG2-PHD) that recognizes histone H3 methylated at lysine 4 (H3K4me) and influences V(D)J recombination. We report here crystal structures of RAG2-PHD alone and complexed with five modified H3 peptides. Two aspects of RAG2-PHD are unique. First, in the absence of the modified peptide, a peptide N-terminal to RAG2-PHD occupies the substrate-binding site, which may reflect an autoregulatory mechanism. Second, in contrast to other H3K4me3-binding PHD domains, RAG2-PHD substitutes a carboxylate that interacts with arginine 2 (R2) with a Tyr, resulting in binding to H3K4me3 that is enhanced rather than inhibited by dimethylation of R2. Five residues involved in histone H3 recognition were found mutated in severe combined immunodeficiency (SCID) patients. Disruption of the RAG2-PHD structure appears to lead to the absence of T and B lymphocytes, whereas failure to bind H3K4me3 is linked to Omenn Syndrome. This work provides a molecular basis for chromatin-dependent gene recombination and presents a single protein domain that simultaneously recognizes two distinct histone modifications, revealing added complexity in the read-out of combinatorial histone modifications.


Subject(s)
DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase/chemistry , Histones/metabolism , Protein Processing, Post-Translational , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , DNA-Binding Proteins/chemistry , Gene Rearrangement , Histones/chemistry , Immunologic Deficiency Syndromes/genetics , Methylation , Mice , Models, Molecular , Molecular Sequence Data , Peptides/metabolism , Protein Binding , Protein Conformation , Protein Interaction Mapping , Protein Structure, Tertiary , Sequence Alignment , Sequence Homology, Amino Acid , Zinc Fingers/physiology
13.
Nature ; 450(7172): 1106-10, 2007 Dec 13.
Article in English | MEDLINE | ID: mdl-18033247

ABSTRACT

Nuclear processes such as transcription, DNA replication and recombination are dynamically regulated by chromatin structure. Eukaryotic transcription is known to be regulated by chromatin-associated proteins containing conserved protein domains that specifically recognize distinct covalent post-translational modifications on histones. However, it has been unclear whether similar mechanisms are involved in mammalian DNA recombination. Here we show that RAG2--an essential component of the RAG1/2 V(D)J recombinase, which mediates antigen-receptor gene assembly--contains a plant homeodomain (PHD) finger that specifically recognizes histone H3 trimethylated at lysine 4 (H3K4me3). The high-resolution crystal structure of the mouse RAG2 PHD finger bound to H3K4me3 reveals the molecular basis of H3K4me3-recognition by RAG2. Mutations that abrogate RAG2's recognition of H3K4me3 severely impair V(D)J recombination in vivo. Reducing the level of H3K4me3 similarly leads to a decrease in V(D)J recombination in vivo. Notably, a conserved tryptophan residue (W453) that constitutes a key structural component of the K4me3-binding surface and is essential for RAG2's recognition of H3K4me3 is mutated in patients with immunodeficiency syndromes. Together, our results identify a new function for histone methylation in mammalian DNA recombination. Furthermore, our results provide the first evidence indicating that disrupting the read-out of histone modifications can cause an inherited human disease.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Gene Rearrangement, B-Lymphocyte , Histones/metabolism , Lysine/metabolism , Recombination, Genetic , VDJ Recombinases/metabolism , Amino Acid Motifs , Animals , Binding Sites , Histones/chemistry , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Humans , Immunologic Deficiency Syndromes/genetics , Lysine/chemistry , Methylation , Mice , Models, Molecular , Protein Binding , Structure-Activity Relationship , Substrate Specificity , Tryptophan/genetics , Tryptophan/metabolism , VDJ Recombinases/chemistry
14.
J Biol Chem ; 280(31): 28701-10, 2005 Aug 05.
Article in English | MEDLINE | ID: mdl-15964836

ABSTRACT

The RAG1 and RAG2 proteins catalyze V(D)J recombination and are essential for generation of the diverse repertoire of antigen receptor genes and effective immune responses. RAG2 is composed of a "core" domain that is required for the recombination reaction and a C-terminal nonessential or "non-core" region. Recent evidence has emerged arguing that the non-core region plays a critical regulatory role in the recombination reaction, and mutations in this region have been identified in patients with immunodeficiencies. Here we present the first structural data for the RAG2 protein, using NMR spectroscopy to demonstrate that the C terminus of RAG2 contains a noncanonical PHD finger. All of the non-core mutations of RAG2 that are implicated in the development of immunodeficiencies are located within the PHD finger, at either zinc-coordinating residues or residues adjacent to an alpha-helix on the surface of the domain that participates in binding to the signaling molecules, phosphoinositides. Functional analysis of disease and phosphoinositide-binding mutations reveals novel intramolecular interactions within the non-core region and suggests that the PHD finger adopts two distinct states. We propose a model in which the equilibrium between these states modulates recombination activity. Together, these data identify the PHD finger as a novel and functionally important domain of RAG2.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Recombination, Genetic , VDJ Recombinases/metabolism , Zinc Fingers , Amino Acid Sequence , Animals , Binding Sites , Consensus Sequence , Kinetics , Mice , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Alignment , Sequence Homology, Amino Acid
15.
Mol Cell Biol ; 25(11): 4413-25, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15899848

ABSTRACT

Efficient assembly of RAG1/2-recombination signal sequence (RSS) DNA complexes that are competent for V(D)J cleavage requires the presence of the nonspecific DNA binding and bending protein HMGB1 or HMGB2. We find that either of the two minimal DNA binding domains of HMGB1 is effective in assembling RAG1/2-RSS complexes on naked DNA and stimulating V(D)J cleavage but that both domains are required for efficient activity when the RSS is incorporated into a nucleosome. The single-domain HMGB protein from Saccharomyces cerevisiae, Nhp6A, efficiently assembles RAG1/2 complexes on naked DNA; however, these complexes are minimally competent for V(D)J cleavage. Nhp6A forms much more stable DNA complexes than HMGB1, and a variety of mutations that destabilize Nhp6A binding to bent microcircular DNA promote increased V(D)J cleavage. One of the two DNA bending wedges on Nhp6A and the analogous phenylalanine wedge at the DNA exit site of HMGB1 domain A were found to be essential for promoting RAG1/2-RSS complex formation. Because the phenylalanine wedge is required for specific recognition of DNA kinks, we propose that HMGB proteins facilitate RAG1/2-RSS interactions by recognizing a distorted DNA structure induced by RAG1/2 binding. The resulting complex must be sufficiently dynamic to enable the series of RAG1/2-mediated chemical reactions on the DNA.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Rearrangement , High Mobility Group Proteins/metabolism , Homeodomain Proteins/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , DNA/metabolism , DNA-Binding Proteins/genetics , HMGB1 Protein , HMGN Proteins , High Mobility Group Proteins/genetics , Homeodomain Proteins/genetics , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phenylalanine/genetics , Phenylalanine/metabolism , Protein Structure, Tertiary , Rats , Repressor Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Deletion , VDJ Recombinases/genetics , VDJ Recombinases/metabolism
16.
J Biol Chem ; 280(21): 20247-52, 2005 May 27.
Article in English | MEDLINE | ID: mdl-15757898

ABSTRACT

Lymphoid cells of the vertebrate immune system rely on factors in the non-homologous end-joining (NHEJ) DNA repair pathway to form signal joints during V(D)J recombination. Unlike other end-joining reactions, signal joint formation is a specialized case of NHEJ that also requires the lymphoid-specific RAG proteins. Whether V(D)J recombination requires the Mre11-Rad50-Nbs1 complex remains an open question, as null mutations in any member of the complex are lethal in mammals. However, Saccharomyces cerevisiae strains carrying null mutations in components of the homologous Mre11p-Rad50p-Xrs2p (MRX) complex are viable. We therefore took advantage of a recently developed V(D)J recombination assay in yeast to assess the role of MRX in V(D)J joining. Here we confirmed that signal joint formation in yeast is dependent on the same NHEJ factors known to be required in mammalian cells. In addition, we showed an absolute requirement for the MRX complex in signal joining, suggesting that the Mre11-Rad50-Nbs1 complex may be required for signal joint formation in mammalian cells as well.


Subject(s)
DNA Repair , DNA-Binding Proteins/physiology , Endodeoxyribonucleases/physiology , Exodeoxyribonucleases/physiology , Saccharomyces cerevisiae Proteins/physiology , VDJ Recombinases/metabolism , DNA Repair/genetics , DNA-Binding Proteins/genetics , Endodeoxyribonucleases/genetics , Exodeoxyribonucleases/genetics , Genes, Immunoglobulin , Genes, T-Cell Receptor , Mutagenesis , Polymerase Chain Reaction , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
18.
Immunol Rev ; 200: 165-81, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15242404

ABSTRACT

Breaking apart chromosomes is not a matter to be taken lightly. The possible negative outcomes are obvious: loss of information, unstable chromosomes, chromosomal translocations, tumorigenesis, or cell death. Utilizing DNA rearrangement to generate the desired diversity in the antigen receptor loci is a risky business, and it must be carefully controlled. In general, the regulation is so precise that the negative consequences are minimal or not apparent. They are visible only when the process of V(D)J recombination goes awry, as for example in some chromosomal translocations associated with lymphoid tumors. Regulation is imposed not only to prevent the generation of random breaks in the DNA, but also to direct rearrangement to the appropriate locus or subregion of a locus in the appropriate cell at the appropriate time. Antigen receptor rearrangement is regulated essentially at four different levels: expression of the RAG1/2 recombinase, intrinsic biochemical properties of the recombinase and the cleavage reaction, the post-cleavage /DNA repair stage of the process, and accessibility of the substrate to the recombinase. Within each of these broad categories, multiple mechanisms are used to achieve the desired aims. The major focus of this review is on accessibility control and the role of chromatin and nuclear architecture in achieving this regulation, although other issues are touched upon.


Subject(s)
Gene Rearrangement, B-Lymphocyte , Gene Rearrangement, T-Lymphocyte , Alleles , Chromatin/chemistry , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Histones/chemistry , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunoglobulin Heavy Chains/immunology , Nuclear Proteins , Nucleosomes/chemistry , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology
19.
J Biol Chem ; 279(34): 35360-7, 2004 Aug 20.
Article in English | MEDLINE | ID: mdl-15201272

ABSTRACT

Control of V(D)J recombination is critical for the generation of a fully developed immune repertoire. The molecular mechanisms underlying the regulation of antigen receptor gene assembly are beginning to be revealed. Here we studied the influence of chromatin modifications on V(D)J cleavage of a polynucleosomal substrate, in which V(D)J cleavage is greatly reduced compared with naked DNA. ATP-dependent remodeling by human SWI/SNF (hSWI/SNF) in the presence of HMG1 led to a substantial increase of cleavage by the recombination activation gene (RAG) proteins. Either BRG1, the ATPase subunit of hSWI/SNF, or SNF2h, the ATPase of human ISWI complexes, was capable of stimulating V(D)J cleavage of the array, although these remodelers act by different mechanisms. No effect of histone hyperacetylation was detectable in this system. As is observed on naked DNA, in the presence of core RAG1, the full-length RAG2 protein proved to be more active than core RAG2 on these polynucleosomal arrays, reinforcing the importance of the RAG2 C-terminal domain for efficient recombination. Comparison of 5 S array cleavage by the RAG proteins or by the restriction enzyme HhaI after remodeling by hSWI/SNF suggested that RAG proteins and HhaI might have different requirements for maximal accessibility of the substrate.


Subject(s)
Adenosine Triphosphatases/genetics , Adenosine Triphosphate/metabolism , Recombination, Genetic , Transcription Factors/genetics , Adenosine Triphosphatases/metabolism , Animals , DNA Helicases , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Escherichia coli , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Transcription Factors/metabolism
20.
EMBO J ; 23(5): 1198-206, 2004 Mar 10.
Article in English | MEDLINE | ID: mdl-14988730

ABSTRACT

Following V(D)J cleavage, the newly liberated DNA signal ends can be either fused together into a signal joint or used as donor DNA in RAG-mediated transposition. We find that both V(D)J cleavage and release of flanking coding DNA occur before the target capture step of transposition can proceed; no coding DNA is ever detected in the target capture complex. Separately from its role in V(D)J cleavage, the DDE motif of the RAG1/2 active site is specifically required for target DNA capture. The requirement for cleavage and release of coding DNA prior to either physical target binding or functional target commitment suggests that the RAG1/2 transposase contains a single binding site for non-RSS DNA that can accommodate either target DNA or coding DNA, but not both together. Perhaps the presence of coding DNA may aid in preventing transpositional resolution of V(D)J recombination intermediates.


Subject(s)
DNA/metabolism , Homeodomain Proteins/metabolism , Recombination, Genetic , Binding Sites , DNA/chemistry , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Nuclear Proteins , Nucleic Acid Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...