Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Article in English | MEDLINE | ID: mdl-34006647

ABSTRACT

Classical nonhomologous end joining (C-NHEJ) repairs DNA double-strand breaks (DSBs) throughout interphase but predominates in G1 phase when homologous recombination is unavailable. Complexes containing the Ku70/80 ("Ku") and XRCC4/ligase IV (Lig4) core C-NHEJ factors are required, respectively, for sensing and joining DSBs. While XRCC4/Lig4 are absolutely required for joining RAG1/2 endonuclease ("RAG")-initiated DSBs during V(D)J recombination in G1-phase progenitor lymphocytes, cycling cells deficient for XRCC4/Lig4 also can join chromosomal DSBs by alternative end-joining (A-EJ) pathways. Restriction of V(D)J recombination by XRCC4/Lig4-mediated joining has been attributed to RAG shepherding V(D)J DSBs exclusively into the C-NHEJ pathway. Here, we report that A-EJ of DSB ends generated by RAG1/2, Cas9:gRNA, and Zinc finger endonucleases in Lig4-deficient G1-arrested progenitor B cell lines is suppressed by Ku. Thus, while diverse DSBs remain largely as free broken ends in Lig4-deficient G1-arrested progenitor B cells, deletion of Ku70 increases DSB rejoining and translocation levels to those observed in Ku70-deficient counterparts. Correspondingly, while RAG-initiated V(D)J DSB joining is abrogated in Lig4-deficient G1-arrested progenitor B cell lines, joining of RAG-generated DSBs in Ku70-deficient and Ku70/Lig4 double-deficient lines occurs through a translocation-like A-EJ mechanism. Thus, in G1-arrested, Lig4-deficient progenitor B cells are functionally end-joining suppressed due to Ku-dependent blockage of A-EJ, potentially in association with G1-phase down-regulation of Lig1. Finally, we suggest that differential impacts of Ku deficiency versus Lig4 deficiency on V(D)J recombination, neuronal apoptosis, and embryonic development results from Ku-mediated inhibition of A-EJ in the G1 cell cycle phase in Lig4-deficient developing lymphocyte and neuronal cells.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , Ku Autoantigen/genetics , Precursor Cells, B-Lymphoid/metabolism , V(D)J Recombination , Animals , DNA Ligase ATP/genetics , DNA Ligase ATP/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , G1 Phase/genetics , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Ku Autoantigen/metabolism , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Precursor Cells, B-Lymphoid/cytology
2.
Neuron ; 108(1): 209-224.e6, 2020 10 14.
Article in English | MEDLINE | ID: mdl-32827456

ABSTRACT

The representation of odor in olfactory cortex (piriform) is distributive and unstructured and can only be afforded behavioral significance upon learning. We performed 2-photon imaging to examine the representation of odors in piriform and in two downstream areas, the orbitofrontal cortex (OFC) and the medial prefrontal cortex (mPFC), as mice learned olfactory associations. In piriform, we observed that odor responses were largely unchanged during learning. In OFC, 30% of the neurons acquired robust responses to conditioned stimuli (CS+) after learning, and these responses were gated by internal state and task context. Moreover, direct projections from piriform to OFC can be entrained to elicit learned olfactory behavior. CS+ responses in OFC diminished with continued training, whereas persistent representations of both CS+ and CS- odors emerged in mPFC. Optogenetic silencing indicates that these two brain structures function sequentially to consolidate the learning of appetitive associations.


Subject(s)
Appetitive Behavior/physiology , Association Learning/physiology , Neurons/physiology , Odorants , Olfactory Pathways/physiology , Piriform Cortex/physiology , Prefrontal Cortex/physiology , Animals , Conditioning, Classical/physiology , Intravital Microscopy , Mice , Microscopy, Fluorescence, Multiphoton , Optogenetics , Piriform Cortex/cytology , Prefrontal Cortex/cytology
3.
Adv Immunol ; 116: 1-49, 2012.
Article in English | MEDLINE | ID: mdl-23063072

ABSTRACT

Classical nonhomologous end joining (C-NHEJ) is one of the two major known pathways for the repair of DNA double-strand breaks (DSBs) in mammalian cells. Our understanding of C-NHEJ has been derived, in significant part, through studies of programmed physiologic DNA DSBs formed during V(D)J recombination in the developing immune system. Studies of immunoglobulin heavy-chain (IgH) class-switch recombination (CSR) also have revealed that there is an "alternative" end-joining process (A-EJ) that can function, relatively robustly, in the repair of DSBs in activated mature B lymphocytes. This A-EJ process has also been implicated in the formation of oncogenic translocations found in lymphoid tumors. In this review, we discuss our current understanding of C-NHEJ and A-EJ in the context of V(D)J recombination, CSR, and the formation of chromosomal translocations.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA Repair , Lymphocytes/metabolism , Animals , Humans , Immunoglobulin Class Switching , V(D)J Recombination
4.
Proc Natl Acad Sci U S A ; 109(7): 2473-8, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22308491

ABSTRACT

Classical nonhomologous DNA end-joining (C-NHEJ), which is a major DNA double-strand break (DSB) repair pathway in mammalian cells, plays a dominant role in joining DSBs during Ig heavy chain (IgH) class switch recombination (CSR) in activated B lymphocytes. However, in B cells deficient for one or more requisite C-NHEJ factors, such as DNA ligase 4 (Lig4) or XRCC4, end-joining during CSR occurs by a distinct alternative end-joining (A-EJ) pathway. A-EJ also has been implicated in joining DSBs found in oncogenic chromosomal translocations. DNA ligase 3 (Lig3) and its cofactor XRCC1 are widely considered to be requisite A-EJ factors, based on biochemical studies or extrachromosomal substrate end-joining studies. However, potential roles for these factors in A-EJ of endogenous chromosomal DSBs have not been tested. Here, we report that Xrcc1 inactivation via conditional gene-targeted deletion in WT or XRCC4-deficient primary B cells does not have an impact on either CSR or IgH/c-myc translocations in activated B lymphocytes. Indeed, homozygous deletion of Xrcc1 does not impair A-EJ of I-SceI-induced DSBs in XRCC4-deficient pro-B-cell lines. Correspondingly, substantial depletion of Lig3 in Lig4-deficient primary B cells or B-cell lines does not impair A-EJ of CSR-mediated DSBs or formation of IgH/c-myc translocations. Our findings firmly demonstrate that XRCC1 is not a requisite factor for A-EJ of chromosomal DSBs and raise the possibility that DNA ligase 1 (Lig1) may contribute more to A-EJ than previously considered.


Subject(s)
DNA Repair , DNA-Binding Proteins/physiology , Animals , B-Lymphocytes/cytology , Cell Lineage , Mice , Translocation, Genetic , X-ray Repair Cross Complementing Protein 1
5.
J Exp Med ; 208(13): 2733-46, 2011 Dec 19.
Article in English | MEDLINE | ID: mdl-22143888

ABSTRACT

Immunoglobulin heavy chain (IgH) class-switch recombination (CSR) replaces initially expressed Cµ (IgM) constant regions (C(H)) exons with downstream C(H) exons. Stimulation of B cells with anti-CD40 plus interleukin-4 induces CSR from Cµ to Cγ1 (IgG1) and Cε (IgE), the latter of which contributes to the pathogenesis of atopic diseases. Although Cε CSR can occur directly from Cµ, most mature peripheral B cells undergo CSR to Cε indirectly, namely from Cµ to Cγ1, and subsequently to Cε. Physiological mechanisms that influence CSR to Cγ1 versus Cε are incompletely understood. In this study, we report a role for B cell developmental maturity in IgE CSR. Based in part on a novel flow cytometric IgE CSR assay, we show that immature B cells preferentially switch to IgE versus IgG1 through a mechanism involving increased direct CSR from Cµ to Cε. Our findings suggest that IgE dysregulation in certain immunodeficiencies may be related to impaired B cell maturation.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin Class Switching/physiology , Immunoglobulin E/immunology , Immunoglobulin epsilon-Chains/immunology , Immunoglobulin mu-Chains/immunology , Recombination, Genetic/physiology , Animals , Common Variable Immunodeficiency/genetics , Common Variable Immunodeficiency/immunology , Immunoglobulin E/genetics , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin epsilon-Chains/genetics , Immunoglobulin mu-Chains/genetics , Mice , Mice, Knockout
6.
Nature ; 469(7329): 250-4, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-21160472

ABSTRACT

Classical non-homologous DNA end-joining (NHEJ) is a major mammalian DNA double-strand-break (DSB) repair pathway. Deficiencies for classical NHEJ factors, such as XRCC4, abrogate lymphocyte development, owing to a strict requirement for classical NHEJ to join V(D)J recombination DSB intermediates. The XRCC4-like factor (XLF; also called NHEJ1) is mutated in certain immunodeficient human patients and has been implicated in classical NHEJ; however, XLF-deficient mice have relatively normal lymphocyte development and their lymphocytes support normal V(D)J recombination. The ataxia telangiectasia-mutated protein (ATM) detects DSBs and activates DSB responses by phosphorylating substrates including histone H2AX. However, ATM deficiency causes only modest V(D)J recombination and lymphocyte developmental defects, and H2AX deficiency does not have a measurable impact on these processes. Here we show that XLF, ATM and H2AX all have fundamental roles in processing and joining DNA ends during V(D)J recombination, but that these roles have been masked by unanticipated functional redundancies. Thus, combined deficiency of ATM and XLF nearly blocks mouse lymphocyte development due to an inability to process and join chromosomal V(D)J recombination DSB intermediates. Combined XLF and ATM deficiency also severely impairs classical NHEJ, but not alternative end-joining, during IgH class switch recombination. Redundant ATM and XLF functions in classical NHEJ are mediated by ATM kinase activity and are not required for extra-chromosomal V(D)J recombination, indicating a role for chromatin-associated ATM substrates. Correspondingly, conditional H2AX inactivation in XLF-deficient pro-B lines leads to V(D)J recombination defects associated with marked degradation of unjoined V(D)J ends, revealing that H2AX has a role in this process.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins/metabolism , Gene Rearrangement, B-Lymphocyte , Histones/metabolism , Protein Serine-Threonine Kinases/metabolism , Recombination, Genetic , Tumor Suppressor Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Line, Transformed , Chromatin/metabolism , Chromosomes, Mammalian/genetics , Chromosomes, Mammalian/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Gene Rearrangement, B-Lymphocyte/genetics , Mice , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
7.
Adv Immunol ; 106: 93-133, 2010.
Article in English | MEDLINE | ID: mdl-20728025

ABSTRACT

Recurrent chromosomal abnormalities, especially chromosomal translocations, are strongly associated with certain subtypes of leukemia, lymphoma and solid tumors. The appearance of particular translocations or associated genomic alterations can be important indicators of disease prognosis, and in some cases, certain translocations may indicate appropriate therapy protocols. To date, most of our knowledge about chromosomal translocations has derived from characterization of the highly selected recurrent translocations found in certain cancers. Until recently, mechanisms that promote or suppress chromosomal translocations, in particular, those responsible for their initiation, have not been addressed. For translocations to occur, two distinct chromosomal loci must be broken, brought together (synapsed) and joined. Here, we discuss recent findings on processes and pathways that influence the initiation of chromosomal translocations, including the generation fo DNA double strand breaks (DSBs) by general factors or in the context of the Lymphocyte-specific V(D)J and IgH class-switch recombination processes. We also discuss the role of spatial proximity of DSBs in the interphase nucleus with respect to how DSBs on different chromosomes are justaposed for joining. In addition, we discuss the DNA DSB response and its role in recognizing and tethering chromosomal DSBs to prevent translocations, as well as potential roles of the classical and alternative DSB end-joining pathways in suppressing or promoting translocations. Finally, we discuss the potential roles of long range regulatory elements, such as the 3'IgH enhancer complex, in promoting the expression of certain translocations that are frequent in lymphomas and, thereby, contributing to their frequent appearance in tumors.


Subject(s)
Neoplasms/genetics , Translocation, Genetic , Animals , B-Lymphocytes/physiology , Chromosome Breakage , DNA Breaks, Double-Stranded , DNA Repair , Gene Expression Regulation, Neoplastic , Genes, RAG-1 , Humans , Immunoglobulin Class Switching , Leukemia/genetics , Lymphoma/genetics , Philadelphia Chromosome , Recombination, Genetic , Regulatory Sequences, Nucleic Acid , T-Lymphocytes/physiology
8.
J Exp Med ; 207(2): 417-27, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20142431

ABSTRACT

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


Subject(s)
Antigens, Nuclear/metabolism , B-Lymphocytes/metabolism , DNA Breaks, Double-Stranded , DNA Ligases/metabolism , DNA Repair , DNA-Binding Proteins/metabolism , Immunoglobulin Class Switching , Immunoglobulins/genetics , Animals , Antigens, Nuclear/immunology , B-Lymphocytes/immunology , DNA Ligase ATP , DNA Ligases/immunology , DNA-Binding Proteins/immunology , Immunoglobulins/metabolism , Ku Autoantigen , Mice
9.
Proc Natl Acad Sci U S A ; 107(7): 3040-5, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20133637

ABSTRACT

Ig heavy chain (IgH) class-switch recombination (CSR) replaces the IgH C mu constant region exons with one of several sets of downstream IgH constant region exons (e.g., C gamma, C epsilon, or C alpha), which affects switching from IgM to another IgH class (e.g., IgG, IgE, or IgA). Activation-induced cytidine deaminase (AID) initiates CSR by promoting DNA double-strand breaks (DSBs) within switch (S) regions flanking the donor C mu (S mu) and a downstream acceptor C(H) (e.g., S gamma, S epsilon, S alpha) that are then joined to complete CSR. DSBs generated in S mu frequently are joined within S mu to form internal switch region deletions (ISD). AID-induced ISD and mutations have been considered rare in downstream S regions, suggesting that AID targeting to these S regions requires its prior recruitment to S mu. We have now assayed for CSR and ISD in B cells lacking S mu (S mu(-/-) B cells). In S mu(-/-) B cells activated for CSR to IgG1 and IgE, CSR to IgG1 was greatly reduced; but, surprisingly, CSR to IgE occurred at nearly normal levels. Moreover, normal B cells had substantial S gamma1 ISD and increased mutations in and near S gamma1, and levels of both were greatly increased in S mu(-/-) B cells. Finally, S mu(-/-) B cells underwent downstream CSR between S gamma1 and S epsilon on alleles that lacked S mu CSR to these sequences. Our findings show that AID targets downstream S regions independently of CSR with Smu and implicate an alternative pathway for IgE class switching that involves generation and joining of DSBs within two different downstream S regions before S mu joining.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin Class Switching/immunology , Immunoglobulin E/biosynthesis , Immunoglobulin Heavy Chains/genetics , Animals , Blotting, Southern , Cytidine Deaminase/metabolism , DNA Mutational Analysis , DNA Primers/genetics , Flow Cytometry , Hybridomas/immunology , Hybridomas/metabolism , Immunoglobulin E/immunology , Immunoglobulin Heavy Chains/immunology , Immunoglobulin M/genetics , Immunoglobulin Switch Region/genetics , Mice , Mice, Knockout
10.
Proc Natl Acad Sci U S A ; 107(7): 3034-9, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20133803

ABSTRACT

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


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair/immunology , Immunoglobulin Class Switching/genetics , Immunoglobulin Switch Region/genetics , Translocation, Genetic/genetics , Animals , Antigens, Nuclear/genetics , B-Lymphocytes/immunology , Base Sequence , Blotting, Southern , DNA Ligase ATP , DNA Ligases/genetics , DNA Primers/genetics , DNA-Binding Proteins/genetics , Genes, myc/genetics , Immunoglobulin Class Switching/immunology , Immunoglobulin Heavy Chains/genetics , In Situ Hybridization, Fluorescence , Ku Autoantigen , Mice , Mice, Knockout , Molecular Sequence Data , Translocation, Genetic/immunology
12.
J Exp Med ; 206(1): 125-38, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19103880

ABSTRACT

We show that the enzymatic acetylation and deacetylation of a cell surface carbohydrate controls B cell development, signaling, and immunological tolerance. Mice with a mutation in sialate:O-acetyl esterase, an enzyme that specifically removes acetyl moieties from the 9-OH position of alpha2-6-linked sialic acid, exhibit enhanced B cell receptor (BCR) activation, defects in peripheral B cell development, and spontaneously develop antichromatin autoantibodies and glomerular immune complex deposits. The 9-O-acetylation state of sialic acid regulates the function of CD22, a Siglec that functions in vivo as an inhibitor of BCR signaling. These results describe a novel catalytic regulator of B cell signaling and underscore the crucial role of inhibitory signaling in the maintenance of immunological tolerance in the B lineage.


Subject(s)
B-Lymphocytes/metabolism , Carboxylic Ester Hydrolases/metabolism , Cell Differentiation/physiology , Receptors, Antigen, B-Cell/physiology , Signal Transduction/physiology , Acetylation , Acetylesterase , Animals , Antibodies, Antinuclear/blood , B-Lymphocyte Subsets/cytology , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Blotting, Western , Bone Marrow Cells/cytology , Carboxylic Ester Hydrolases/genetics , Cell Count , Glomerulonephritis/genetics , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Models, Biological , Mutation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sialic Acid Binding Ig-like Lectin 2/metabolism , Sialic Acids/metabolism , Spleen/cytology
13.
J Exp Med ; 205(3): 557-64, 2008 Mar 17.
Article in English | MEDLINE | ID: mdl-18316419

ABSTRACT

The DNA-dependent protein kinase catalytic subunit (DNA-PKcs) and Artemis are classical nonhomologous DNA end-joining (C-NHEJ) factors required for joining a subset of DNA double-strand breaks (DSB), particularly those requiring end processing. In mature B cells, activation-induced cytidine deaminase (AID) initiates class switch recombination (CSR) by introducing lesions into S regions upstream of two recombining C(H) exons, which are processed into DSBs and rejoined by C-NHEJ to complete CSR. The function of DNA-PKcs in CSR has been controversial with some reports but not others showing that DNA-PKcs-deficient mice are significantly impaired for CSR. Artemis-deficient B cells reportedly undergo CSR at normal levels. Overall, it is still not known whether there are any CSR-associated DSBs that require DNA-PKcs and/or Artemis to be joined. Here, we have used an immunoglobulin (Ig)H locus-specific fluorescent in situ hybridization assay to unequivocally demonstrate that both DNA-PKcs and, unexpectedly, Artemis are necessary for joining a subset of AID-dependent DSBs. In the absence of either factor, B cells activated for CSR frequently generate AID-dependent IgH locus chromosomal breaks and translocations. We also find that under specific activation conditions, DNA-PKcs(-/-) B cells with chromosomal breaks are eliminated or at least prevented from progressing to metaphase via a p53-dependent response.


Subject(s)
DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Immunoglobulin Class Switching , Nuclear Proteins/metabolism , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Base Sequence , DNA Probes/genetics , DNA Repair , DNA-Activated Protein Kinase/deficiency , DNA-Activated Protein Kinase/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Endonucleases , Genomic Instability , Immunoglobulin Heavy Chains/genetics , In Situ Hybridization, Fluorescence , Mice , Mice, Knockout , Mice, Transgenic , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Recombination, Genetic , Translocation, Genetic
14.
Nature ; 449(7161): 478-82, 2007 Sep 27.
Article in English | MEDLINE | ID: mdl-17713479

ABSTRACT

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


Subject(s)
Genes, Immunoglobulin Heavy Chain/genetics , Immunoglobulin Class Switching/genetics , Recombination, Genetic/genetics , Translocation, Genetic/genetics , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , B-Lymphocytes/radiation effects , Base Sequence , Cell Proliferation , Cells, Cultured , Chromosome Breakage , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , In Situ Hybridization, Fluorescence , Mice , Radiation, Ionizing , Telomere/genetics
15.
J Immunol ; 179(4): 2270-81, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17675488

ABSTRACT

Disparate models for the development of peripheral B cells may reflect significant heterogeneity in recirculating long-lived B cells that have not been previously accounted for. We show in this study that the murine recirculating B cell pool contains two distinct, long-lived, posttransitional, follicular B cell populations. Follicular Type I IgM(low) B cells require Ag-derived and Btk-dependent signals for their development and make up the majority of cells in the recirculating follicular B cell pool. Follicular type II B cells do not require Btk- or Notch-2-derived signals, make up about a third of the long-lived recirculating B cell pool, and can develop in the absence of Ag. These two follicular populations exhibit differences in basal tyrosine phosphorylation and in BCR-induced proliferation, suggesting that they may represent functionally distinct populations of long-lived recirculating B cells.


Subject(s)
B-Lymphocytes/immunology , Cell Proliferation , Immunoglobulin M/immunology , Protein-Tyrosine Kinases/immunology , Receptor, Notch2/immunology , Receptors, Antigen, B-Cell/immunology , Agammaglobulinaemia Tyrosine Kinase , Animals , B-Lymphocytes/cytology , Mice , Phosphorylation , Signal Transduction/immunology
16.
J Immunol ; 179(1): 195-200, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17579038

ABSTRACT

NF-kappaB1 and Notch2 are both required for the development of marginal zone (MZ) B cells. Analysis of B lymphocyte development in mice that are doubly heterozygous at the Notch2 and NF-kappaB1 loci revealed synergism between Notch2 and NF-kappaB1 during MZ B cell development. Two known transcriptional targets of the Notch pathway, Hes-5 and Deltex-1, were found to be preferentially expressed in MZ B cells and regulated by NF-kappaB1. These studies provide in vivo evidence for a genetic interaction between the Notch and NF-kappaB pathways.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Cell Communication/immunology , Cell Differentiation/immunology , NF-kappa B p50 Subunit/physiology , Receptor, Notch2/physiology , Spleen/immunology , Spleen/metabolism , Agammaglobulinaemia Tyrosine Kinase , Animals , B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/pathology , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Communication/genetics , Cell Differentiation/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Enzyme Activation/immunology , Gene Expression Regulation/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , NF-kappa B p50 Subunit/deficiency , NF-kappa B p50 Subunit/genetics , Protein-Tyrosine Kinases/metabolism , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Signal Transduction/immunology , Spleen/pathology , Ubiquitin-Protein Ligases
17.
Mol Immunol ; 43(10): 1694-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16256200

ABSTRACT

Protein kinase C-associated kinase (PKK; DIK/RIP4) is an ankyrin-repeat containing serine/threonine receptor-interacting protein (RIP)-family kinase that can activate NFkappaB, and is required for keratinocyte development. In earlier studies, the expression of a catalytically inactive mutant of PKK in the B cell lineage resulted in a marked decrease in peripheral B cells in the spleen and a severe reduction of B-1 B cells. Here we explore the consequences of a null mutation in PKK with respect to the generation of peripheral B cell lineages and the activation of NFkappaB. We show that PKK is not required for the production of B cells in the bone marrow or for the development and maintenance of all mature B lymphocyte populations. We also show that PKK is not required for the activation of NFkappaB downstream of the BCR, CD40, or TLR-4 in B cells. Taken together, these data demonstrate that the loss of this RIP-family kinase does not compromise B lymphocyte development and maintenance, but leaves open the possibility that PKK may have a redundant role in these processes.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/enzymology , Protein Kinases/physiology , Animals , Bone Marrow/immunology , CD40 Antigens/metabolism , Cell Lineage , Enzyme Activation , Mice , Mice, Mutant Strains , Mutation , Protein Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Antigen, B-Cell/metabolism , Toll-Like Receptor 4/metabolism , NF-kappaB-Inducing Kinase
18.
J Immunol ; 172(9): 5478-88, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15100289

ABSTRACT

The addition of nontemplated (N) nucleotides to coding ends in V(D)J recombination is the result of the action of a unique DNA polymerase, TdT. Although N-nucleotide addition by TdT plays a critical role in the generation of a diverse repertoire of Ag receptor genes, the mechanism by which TdT acts remains unclear. We conducted a structure-function analysis of the murine TdT protein to determine the roles of individual structural motifs that have been implicated in protein-protein and protein-DNA interactions important for TdT function in vivo. This analysis demonstrates that the N-terminal portion of TdT, including the BRCA-1 C-terminal (BRCT) domain, is not required for TdT activity, although the BRCT domain clearly contributes quantitatively to N-nucleotide addition activity. The second helix-hairpin-helix domain of TdT, but not the first, is required for activity. Deletional analysis also suggested that the entire C-terminal region of TdT is necessary for N-nucleotide addition in vivo. The long isoform of TdT was found to reduce N-nucleotide addition by the short form of TdT, but did not increase nucleotide deletion from coding ends in either human or rodent nonlymphoid cells. We consider these results in light of the recently reported structure of the catalytic region of TdT.


Subject(s)
Antibody Diversity/genetics , DNA Nucleotidylexotransferase/metabolism , Immunoglobulin Joining Region/genetics , Immunoglobulin Variable Region/genetics , Nucleotides/metabolism , Recombination, Genetic/immunology , Animals , CHO Cells , Cell Line , Cricetinae , DNA Mutational Analysis , DNA Nucleotidylexotransferase/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Rearrangement/immunology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunoglobulin Joining Region/metabolism , Immunoglobulin Variable Region/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Nuclear Proteins , Nucleotides/genetics , Open Reading Frames/genetics , Plasmids/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Substrate Specificity/genetics , Templates, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...