Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 14 de 14
Filtrer
1.
PLoS Genet ; 11(9): e1005517, 2015 Sep.
Article de Anglais | MEDLINE | ID: mdl-26368543

RÉSUMÉ

Phosphorylation of histone H2AX by ATM and ATR establishes a chromatin recruitment platform for DNA damage response proteins. Phospho-H2AX (γH2AX) has been most intensively studied in the context of DNA double-strand breaks caused by exogenous clastogens, but recent studies suggest that DNA replication stress also triggers formation of γH2A (ortholog of γH2AX) in Schizosaccharomyces pombe. Here, a focused genetic screen in fission yeast reveals that γH2A is critical when there are defects in Replication Factor C (RFC), which loads proliferating cell nuclear antigen (PCNA) clamp onto duplex DNA. Surprisingly Chk1, Cds1/Chk2 and the Rad9-Hus1-Rad1 checkpoint clamp, which are crucial for surviving many genotoxins, are fully dispensable in RFC-defective cells. Immunoblot analysis confirms that Rad9-Hus1-Rad1 is not required for formation of γH2A by Rad3/ATR in S-phase. Defects in DNA polymerase epsilon, which binds PCNA in the replisome, also create an acute need for γH2A. These requirements for γH2A were traced to its role in docking with Brc1, which is a 6-BRCT-domain protein that is structurally related to budding yeast Rtt107 and mammalian PTIP. Brc1, which localizes at stalled replication forks by binding γH2A, prevents aberrant formation of Replication Protein A (RPA) foci in RFC-impaired cells, suggesting that Brc1-coated chromatin stabilizes replisomes when PCNA or DNA polymerase availability limits DNA synthesis.


Sujet(s)
Histone/métabolisme , Phase S , Schizosaccharomyces/métabolisme , DNA polymerase II/génétique , DNA polymerase II/métabolisme , ADN fongique/métabolisme , Mutation , Antigène nucléaire de prolifération cellulaire/métabolisme , Liaison aux protéines , Schizosaccharomyces/cytologie , Schizosaccharomyces/génétique , Protéines de Schizosaccharomyces pombe/génétique , Protéines de Schizosaccharomyces pombe/métabolisme
2.
J Exp Med ; 210(1): 23-30, 2013 Jan 14.
Article de Anglais | MEDLINE | ID: mdl-23267013

RÉSUMÉ

B cell development requires tight regulation to allow for the generation of a diverse repertoire while preventing the development of autoreactive cells. We report, using N-ethyl-N-nitrosourea (ENU)-induced mutagenesis, the identification of a mutant mouse (chompB) with a block in early B cell development. The blockade occurs after the transitional 1 (T1) stage and leads to a decrease in mature B cell subsets and deficits in T cell-dependent antibody responses. Additionally, chompB mice have decreases in myeloid dendritic cells (DCs). The mutation was mapped to the intramembrane protease signal peptide peptidase-like 2a (Sppl2a), a gene not previously implicated in immune cell development. Proteomic analysis identified the invariant chain (CD74) as a key substrate of Sppl2a and suggests that regulated intramembrane proteolysis of CD74 by Sppl2a contributes to B cell and DC survival. Moreover, these data suggest that modulation of Sppl2a may be a useful therapeutic strategy for treatment of B cell dependent autoimmune disorders.


Sujet(s)
Aspartic acid endopeptidases/métabolisme , Lymphocytes B/physiologie , Cellules dendritiques/anatomopathologie , Protéines membranaires/métabolisme , Animaux , Antigènes de différenciation des lymphocytes B/génétique , Antigènes de différenciation des lymphocytes B/métabolisme , Aspartic acid endopeptidases/génétique , Lymphocytes B/anatomopathologie , Survie cellulaire , Cellules dendritiques/physiologie , 1-Éthyl-1-nitroso-urée/pharmacologie , Antigènes d'histocompatibilité de classe II/génétique , Antigènes d'histocompatibilité de classe II/métabolisme , Immunoglobulines/métabolisme , Activation des lymphocytes , Protéines membranaires/génétique , Souris , Souches mutantes de souris , Mutagenèse/effets des médicaments et des substances chimiques , Mutation , Lymphocytes T/immunologie , Lymphocytes T/métabolisme
3.
Philos Trans R Soc Lond B Biol Sci ; 366(1584): 3562-71, 2011 Dec 27.
Article de Anglais | MEDLINE | ID: mdl-22084383

RÉSUMÉ

Double-strand breaks (DSBs), arising from exposure to exogenous clastogens or as a by-product of endogenous cellular metabolism, pose grave threats to genome integrity. DSBs can sever whole chromosomes, leading to chromosomal instability, a hallmark of cancer. Healing broken DNA takes time, and it is therefore essential to temporarily halt cell division while DSB repair is underway. The seminal discovery of cyclin-dependent kinases as master regulators of the cell cycle unleashed a series of studies aimed at defining how the DNA damage response network delays cell division. These efforts culminated with the identification of Cdc25, the protein phosphatase that activates Cdc2/Cdk1, as a critical target of the checkpoint kinase Chk1. However, regulation works both ways, as recent studies have revealed that Cdc2 activity and cell cycle position determine whether DSBs are repaired by non-homologous end-joining or homologous recombination (HR). Central to this regulation are the proteins that initiate the processing of DNA ends for HR repair, Mre11-Rad50-Nbs1 protein complex and Ctp1/Sae2/CtIP, and the checkpoint kinases Tel1/ATM and Rad3/ATR. Here, we review recent findings and provide insight on how proteins that regulate cell cycle progression affect DSB repair, and, conversely how proteins that repair DSBs affect cell cycle progression.


Sujet(s)
Points de contrôle du cycle cellulaire , Protéines du cycle cellulaire/métabolisme , Cassures double-brin de l'ADN , Réparation de l'ADN par jonction d'extrémités , Réparation de l'ADN par recombinaison , Animaux , Protéines du cycle cellulaire/génétique , Checkpoint kinase 1 , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines fongiques/génétique , Protéines fongiques/métabolisme , Réseaux de régulation génique , Mammifères/génétique , Mammifères/métabolisme , Modèles génétiques , Protein kinases/génétique , Protein kinases/métabolisme , Levures/génétique , Levures/métabolisme , cdc25 Phosphatases/génétique , cdc25 Phosphatases/métabolisme
4.
DNA Repair (Amst) ; 10(10): 1051-9, 2011 Oct 10.
Article de Anglais | MEDLINE | ID: mdl-21889916

RÉSUMÉ

The generation of high affinity antibodies in B cells critically depends on translesion synthesis (TLS) polymerases that introduce mutations into immunoglobulin genes during somatic hypermutation (SHM). The majority of mutations at A/T base pairs during SHM require ubiquitination of PCNA at lysine 164 (PCNA-Ub), which activates TLS polymerases. By comparing the mutation spectra in B cells of WT, TLS polymerase η (Polη)-deficient, PCNA(K164R)-mutant, and PCNA(K164R);Polη double-mutant mice, we now find that most PCNA-Ub-independent A/T mutagenesis during SHM is mediated by Polη. In addition, upon exposure to various DNA damaging agents, PCNA(K164R) mutant cells display strongly impaired recruitment of TLS polymerases, reduced daughter strand maturation and hypersensitivity. Interestingly, compared to the single mutants, PCNA(K164R);Polη double-mutant cells are dramatically delayed in S phase progression and far more prone to cell death following UV exposure. Taken together, these data support the existence of PCNA ubiquitination-dependent and -independent activation pathways of Polη during SHM and DNA damage tolerance.


Sujet(s)
Lymphocytes B/métabolisme , Altération de l'ADN/génétique , Réparation de l'ADN/génétique , DNA-directed DNA polymerase/métabolisme , Antigène nucléaire de prolifération cellulaire/métabolisme , Hypermutation somatique des gènes des immunoglobulines/génétique , Ubiquitination , Animaux , Lymphocytes B/cytologie , Activation enzymatique , Lysine/génétique , Souris , Souris de lignée C57BL , Mutagenèse , Mutation , Antigène nucléaire de prolifération cellulaire/génétique , Rayons ultraviolets
5.
PLoS Genet ; 7(9): e1002262, 2011 Sep.
Article de Anglais | MEDLINE | ID: mdl-21931560

RÉSUMÉ

Translesion DNA synthesis (TLS) is a DNA damage tolerance mechanism in which specialized low-fidelity DNA polymerases bypass replication-blocking lesions, and it is usually associated with mutagenesis. In Saccharomyces cerevisiae a key event in TLS is the monoubiquitination of PCNA, which enables recruitment of the specialized polymerases to the damaged site through their ubiquitin-binding domain. In mammals, however, there is a debate on the requirement for ubiquitinated PCNA (PCNA-Ub) in TLS. We show that UV-induced Rpa foci, indicative of single-stranded DNA (ssDNA) regions caused by UV, accumulate faster and disappear more slowly in Pcna(K164R/K164R) cells, which are resistant to PCNA ubiquitination, compared to Pcna(+/+) cells, consistent with a TLS defect. Direct analysis of TLS in these cells, using gapped plasmids with site-specific lesions, showed that TLS is strongly reduced across UV lesions and the cisplatin-induced intrastrand GG crosslink. A similar effect was obtained in cells lacking Rad18, the E3 ubiquitin ligase which monoubiquitinates PCNA. Consistently, cells lacking Usp1, the enzyme that de-ubiquitinates PCNA exhibited increased TLS across a UV lesion and the cisplatin adduct. In contrast, cells lacking the Rad5-homologs Shprh and Hltf, which polyubiquitinate PCNA, exhibited normal TLS. Knocking down the expression of the TLS genes Rev3L, PolH, or Rev1 in Pcna(K164R/K164R) mouse embryo fibroblasts caused each an increased sensitivity to UV radiation, indicating the existence of TLS pathways that are independent of PCNA-Ub. Taken together these results indicate that PCNA-Ub is required for maximal TLS. However, TLS polymerases can be recruited to damaged DNA also in the absence of PCNA-Ub, and perform TLS, albeit at a significantly lower efficiency and altered mutagenic specificity.


Sujet(s)
Réparation de l'ADN , Réplication de l'ADN , ADN/biosynthèse , Antigène nucléaire de prolifération cellulaire/métabolisme , Ubiquitination , Animaux , Cisplatine/pharmacologie , ADN/effets des médicaments et des substances chimiques , ADN/génétique , Altération de l'ADN , ADN simple brin/biosynthèse , ADN simple brin/génétique , Souris , Mutagenèse , Antigène nucléaire de prolifération cellulaire/génétique , Ubiquitine/génétique , Ubiquitine/métabolisme , Rayons ultraviolets
6.
PLoS Genet ; 7(9): e1002271, 2011 Sep.
Article de Anglais | MEDLINE | ID: mdl-21931565

RÉSUMÉ

The multifunctional Mre11-Rad50-Nbs1 (MRN) protein complex recruits ATM/Tel1 checkpoint kinase and CtIP/Ctp1 homologous recombination (HR) repair factor to double-strand breaks (DSBs). HR repair commences with the 5'-to-3' resection of DNA ends, generating 3' single-strand DNA (ssDNA) overhangs that bind Replication Protein A (RPA) complex, followed by Rad51 recombinase. In Saccharomyces cerevisiae, the Mre11-Rad50-Xrs2 (MRX) complex is critical for DSB resection, although the enigmatic ssDNA endonuclease activity of Mre11 and the DNA-end processing factor Sae2 (CtIP/Ctp1 ortholog) are largely unnecessary unless the resection activities of Exo1 and Sgs1-Dna2 are also eliminated. Mre11 nuclease activity and Ctp1/CtIP are essential for DSB repair in Schizosaccharomyces pombe and mammals. To investigate DNA end resection in Schizo. pombe, we adapted an assay that directly measures ssDNA formation at a defined DSB. We found that Mre11 and Ctp1 are essential for the efficient initiation of resection, consistent with their equally crucial roles in DSB repair. Exo1 is largely responsible for extended resection up to 3.1 kb from a DSB, with an activity dependent on Rqh1 (Sgs1) DNA helicase having a minor role. Despite its critical function in DSB repair, Mre11 nuclease activity is not required for resection in fission yeast. However, Mre11 nuclease and Ctp1 are required to disassociate the MRN complex and the Ku70-Ku80 nonhomologous end-joining (NHEJ) complex from DSBs, which is required for efficient RPA localization. Eliminating Ku makes Mre11 nuclease activity dispensable for MRN disassociation and RPA localization, while improving repair of a one-ended DSB formed by replication fork collapse. From these data we propose that release of the MRN complex and Ku from DNA ends by Mre11 nuclease activity and Ctp1 is a critical step required to expose ssDNA for RPA localization and ensuing HR repair.


Sujet(s)
Protéines chromosomiques nonhistones/génétique , Réparation de l'ADN par jonction d'extrémités/génétique , Protéines de liaison à l'ADN/génétique , Protéines de Schizosaccharomyces pombe/génétique , Schizosaccharomyces/génétique , Cassures double-brin de l'ADN , Helicase/génétique , Réparation de l'ADN/génétique , ADN simple brin/génétique , Exodeoxyribonucleases/génétique , Recombinaison homologue/génétique , Complexes multiprotéiques/génétique , Rad51 Recombinase/génétique , Protéine A de réplication/génétique , Schizosaccharomyces/métabolisme
7.
J Exp Med ; 206(12): 2603-11, 2009 Nov 23.
Article de Anglais | MEDLINE | ID: mdl-19901081

RÉSUMÉ

During somatic hypermutation (SHM), B cells introduce mutations into their immunoglobulin genes to generate high affinity antibodies. Current models suggest a separation in the generation of G/C transversions by the Ung2-dependent pathway and the generation of A/T mutations by the Msh2/ubiquitinated proliferating cell nuclear antigen (PCNA-Ub)-dependent pathway. It is currently unknown whether these pathways compete to initiate mutagenesis and whether PCNA-Ub functions downstream of Ung2. Furthermore, these models do not explain why mice lacking Msh2 have a more than twofold reduction in the total mutation frequency. Our data indicate that PCNA-Ub is required for A/T mutagenesis downstream of both Msh2 and Ung2. Furthermore, we provide evidence that both pathways are noncompetitive to initiate mutagenesis and even collaborate to generate half of all G/C transversions. These findings significantly add to our understanding of SHM and necessitate an update of present SHM models.


Sujet(s)
Lymphocytes B/métabolisme , Protéine-2 homologue de MutS/métabolisme , Antigène nucléaire de prolifération cellulaire/métabolisme , Hypermutation somatique des gènes des immunoglobulines/physiologie , Uracil-DNA glycosidase/métabolisme , Animaux , Affinité des anticorps/physiologie , Lymphocytes B/immunologie , Souris , Souches mutantes de souris , Protéine-2 homologue de MutS/génétique , Protéine-2 homologue de MutS/immunologie , Mutation/immunologie , Antigène nucléaire de prolifération cellulaire/génétique , Antigène nucléaire de prolifération cellulaire/immunologie , Protéines ubiquitinées/génétique , Protéines ubiquitinées/immunologie , Protéines ubiquitinées/métabolisme , Uracil-DNA glycosidase/génétique , Uracil-DNA glycosidase/immunologie
8.
Philos Trans R Soc Lond B Biol Sci ; 364(1517): 621-9, 2009 Mar 12.
Article de Anglais | MEDLINE | ID: mdl-19008189

RÉSUMÉ

Proliferating cell nuclear antigen (PCNA) encircles DNA as a ring-shaped homotrimer and, by tethering DNA polymerases to their template, PCNA serves as a critical replication factor. In contrast to high-fidelity DNA polymerases, the activation of low-fidelity translesion synthesis (TLS) DNA polymerases seems to require damage-inducible monoubiquitylation (Ub) of PCNA at lysine residue 164 (PCNA-Ub). TLS polymerases can tolerate DNA damage, i.e. they can replicate across DNA lesions. The lack of proofreading activity, however, renders TLS highly mutagenic. The advantage is that B cells use mutagenic TLS to introduce somatic mutations in immunoglobulin (Ig) genes to generate high-affinity antibodies. Given the critical role of PCNA-Ub in activating TLS and the role of TLS in establishing somatic mutations in immunoglobulin genes, we analysed the mutation spectrum of somatically mutated immunoglobulin genes in B cells from PCNAK164R knock-in mice. A 10-fold reduction in A/T mutations is associated with a compensatory increase in G/C mutations-a phenotype similar to Poleta and mismatch repair-deficient B cells. Mismatch recognition, PCNA-Ub and Poleta probably act within one pathway to establish the majority of mutations at template A/T. Equally relevant, the G/C mutator(s) seems largely independent of PCNAK(164) modification.


Sujet(s)
Réparation de l'ADN , Antigène nucléaire de prolifération cellulaire/génétique , Hypermutation somatique des gènes des immunoglobulines/génétique , Animaux , Analyse de mutations d'ADN , DNA-directed DNA polymerase/métabolisme , Souris , Souches mutantes de souris , Modèles génétiques , Nucleotidyltransferases/métabolisme , Antigène nucléaire de prolifération cellulaire/métabolisme , Ubiquitination
9.
J Exp Med ; 204(8): 1989-98, 2007 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-17664295

RÉSUMÉ

B cells use translesion DNA synthesis (TLS) to introduce somatic mutations around genetic lesions caused by activation-induced cytidine deaminase. Monoubiquitination at lysine(164) of proliferating cell nuclear antigen (PCNA(K164)) stimulates TLS. To determine the role of PCNA(K164) modifications in somatic hypermutation, PCNA(K164R) knock-in mice were generated. PCNA(K164R/K164R) mutants are born at a sub-Mendelian frequency. Although PCNA(K164R/K164R) B cells proliferate and class switch normally, the mutation spectrum of hypermutated immunoglobulin (Ig) genes alters dramatically. A strong reduction of mutations at template A/T is associated with a compensatory increase at G/C, which is a phenotype similar to polymerase eta (Poleta) and mismatch repair-deficient B cells. Mismatch recognition, monoubiquitinated PCNA, and Poleta likely cooperate in establishing mutations at template A/T during replication of Ig genes.


Sujet(s)
Adénine/composition chimique , Cytosine/composition chimique , Régulation de l'expression des gènes , Immunoglobulines/génétique , Mutagenèse , Mutation , Antigène nucléaire de prolifération cellulaire/génétique , Antigène nucléaire de prolifération cellulaire/physiologie , Animaux , Lymphocytes B/métabolisme , Prolifération cellulaire , DNA-directed DNA polymerase/métabolisme , Homozygote , Souris , Modèles biologiques , Phénotype , Ubiquitine/composition chimique , Ubiquitine/métabolisme
10.
Mol Biotechnol ; 34(2): 101-8, 2006 Oct.
Article de Anglais | MEDLINE | ID: mdl-17172655

RÉSUMÉ

Density-dependent growth inhibition secures tissue homeostasis. Dysfunction of the mechanisms, which regulate this type of growth control is a major cause of neoplasia. In confluent normal rat kidney (NRK) fibroblasts, epidermal growth factor (EGF) receptor levels decline, ultimately rendering these cells irresponsive to EGF. Using an activator protein (AP)-1 sensitive reporter construct, we show that AP-1 activity is strongly decreased in density-arrested NRK cells, but is restored after relaxation of densitydependent growth inhibition by removing neighboring cells. EGF could not induce AP-1 activity or S-phase entry in density-arrested cells, but could do so after pretreatment with retinoic acid, which enhances EGF receptor expression. Our results support a model in which the EGF receptor regulates density-dependent growth control in NRK fibroblasts, which is reflected by EGF-induced mitogenic signaling and consequent AP-1 activity.


Sujet(s)
Prolifération cellulaire , Récepteurs ErbB/physiologie , Fibroblastes/cytologie , Facteur de transcription AP-1/métabolisme , Animaux , Cycle cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Facteur de croissance épidermique/pharmacologie , Récepteurs ErbB/agonistes , Fibroblastes/effets des médicaments et des substances chimiques , Rein/cytologie , Mitogen-Activated Protein Kinase Kinases/métabolisme , Rats , Transduction du signal , Facteur de transcription AP-1/agonistes , Trétinoïne/pharmacologie
11.
J Exp Med ; 203(2): 319-23, 2006 Feb 20.
Article de Anglais | MEDLINE | ID: mdl-16476771

RÉSUMÉ

Somatic hypermutation of Ig genes enables B cells of the germinal center to generate high-affinity immunoglobulin variants. Key intermediates in somatic hypermutation are deoxyuridine lesions, introduced by activation-induced cytidine deaminase. These lesions can be processed further to abasic sites by uracil DNA glycosylase. Mutagenic replication of deoxyuridine, or of its abasic derivative, by translesion synthesis polymerases is hypothesized to underlie somatic hypermutation. Rev1 is a translesion synthesis polymerase that in vitro incorporates uniquely deoxycytidine opposite deoxyuridine and abasic residues. To investigate a role of Rev1 in mammalian somatic hypermutation we have generated mice deficient for Rev1. Although Rev1-/- mice display transient growth retardation, proliferation of Rev1-/- LPS-stimulated B cells is indistinguishable from wild-type cells. In mutated Ig genes from Rev1-/- mice, C to G transversions were virtually absent in the nontranscribed (coding) strand and reduced in the transcribed strand. This defect is associated with an increase of A to T, C to A, and T to C substitutions. These results indicate that Rev1 incorporates deoxycytidine residues, most likely opposite abasic nucleotides, during somatic hypermutation. In addition, loss of Rev1 causes compensatory increase in mutagenesis by other translesion synthesis polymerases.


Sujet(s)
Désoxycytidine/génétique , Gènes d'immunoglobuline , Guanine , Nucleotidyltransferases/déficit , Mutation ponctuelle , Hypermutation somatique des gènes des immunoglobulines/génétique , Animaux , Lymphocytes B/enzymologie , Lymphocytes B/métabolisme , DNA-directed DNA polymerase , Désoxycytidine/métabolisme , Guanine/métabolisme , Immunoglobuline D/génétique , Immunoglobuline M/génétique , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Nucleotidyltransferases/génétique , Uracil-DNA glycosidase/génétique , Uracil-DNA glycosidase/métabolisme
12.
Nucleic Acids Res ; 33(22): e188, 2005 Dec 09.
Article de Anglais | MEDLINE | ID: mdl-16340005

RÉSUMÉ

Embryonic stem (ES) cell technology allows modification of the mouse germline from large deletions and insertions to single nucleotide substitutions by homologous recombination. Identification of these rare events demands an accurate and fast detection method. Current methods for detection rely on Southern blotting and/or conventional PCR. Both the techniques have major drawbacks, Southern blotting is time-consuming and PCR can generate false positives. As an alternative, we here demonstrate a novel approach of Multiplex Ligation-dependent Probe Amplification (MLPA) as a quick, quantitative and reliable method for the detection of homologous, non-homologous and incomplete recombination events in ES cell clones. We have adapted MLPA to detect homologous recombinants in ES cell clones targeted with two different constructs: one introduces a single nucleotide change in the PCNA gene and the other allows for a conditional inactivation of the wild-type PCNA allele. By using MLPA probes consisting of three oligonucleotides we were able to simultaneously detect and quantify both wild-type and mutant alleles.


Sujet(s)
Ciblage de gène/méthodes , Souris/génétique , Sondes oligonucléotidiques , Réaction de polymérisation en chaîne/méthodes , Recombinaison génétique , Animaux , Technique de Southern , Clones cellulaires , ADN/analyse , Embryon de mammifère/cytologie , Antigène nucléaire de prolifération cellulaire/génétique , Cellules souches/composition chimique
13.
Nucleic Acids Res ; 33(1): 356-65, 2005.
Article de Anglais | MEDLINE | ID: mdl-15653636

RÉSUMÉ

Rev1 is a deoxycytidyl transferase associated with DNA translesion synthesis (TLS). In addition to its catalytic domain, Rev1 possesses a so-called BRCA1 C-terminal (BRCT) domain. Here, we describe cells and mice containing a targeted deletion of this domain. Rev1(B/B) mice are healthy, fertile and display normal somatic hypermutation. Rev1(B/B) cells display an elevated spontaneous frequency of intragenic deletions at Hprt. In addition, these cells were sensitized to exogenous DNA damages. Ultraviolet-C (UV-C) light induced a delayed progression through late S and G2 phases of the cell cycle and many chromatid aberrations, specifically in a subset of mutant cells, but not enhanced sister chromatid exchanges (SCE). UV-C-induced mutagenesis was reduced and mutations at thymidine-thymidine dimers were absent in Rev1(B/B) cells, the opposite phenotype of UV-C-exposed cells from XP-V patients, lacking TLS polymerase eta. This suggests that the enhanced UV-induced mutagenesis in XP-V patients may depend on error-prone Rev1-dependent TLS. Together, these data indicate a regulatory role of the Rev1 BRCT domain in TLS of a limited spectrum of endogenous and exogenous nucleotide damages during a defined phase of the cell cycle.


Sujet(s)
Altération de l'ADN , Réplication de l'ADN , Nucleotidyltransferases/composition chimique , Animaux , Protéine BRCA1/composition chimique , Cycle cellulaire/effets des radiations , Aberrations des chromosomes , ADN/biosynthèse , DNA-directed DNA polymerase , Embryon de mammifère/cytologie , Souris , Mutagenèse , Nucleotidyltransferases/génétique , Nucleotidyltransferases/métabolisme , Structure tertiaire des protéines , Échange de chromatides soeurs , Cellules souches/cytologie , Cellules souches/effets des radiations , Cellules souches/ultrastructure , Rayons ultraviolets
14.
J Immunol ; 172(12): 7432-41, 2004 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-15187121

RÉSUMÉ

Studies on human B cells have featured CD27 as a marker and mediator of the B cell response. We have studied CD27 expression and function on B cells in the mouse. We find that B cells acquire CD27 at the centroblast stage and lose it progressively upon further differentiation. It is not a marker for somatically mutated B cells and is present at very low frequency on memory B cells. Enrichment of CD27 among centroblasts and the presence of its ligand CD70 on occasional T and B cells in or near germinal centers (GCs) suggested a role for CD27/CD70 interactions in clonal B cell expansion. Accordingly, GC formation in response to influenza virus infection was delayed in CD27 knockout mice. CD27 deficiency did not affect somatic hypermutation or serum levels of virus-specific IgM, IgG, and IgA attained in primary and recall responses. Adoptive transfer of T and B cells into CD27/CD28(-/-) mice revealed that CD27 promotes GC formation and consequent IgG production by two distinct mechanisms. Stimulation of CD27 on B cells by CD28(+) Th cells accelerates GC formation, most likely by promoting centroblast expansion. In addition, CD27 on T cells can partially substitute for CD28 in supporting GC formation.


Sujet(s)
Lymphocytes B/immunologie , Centre germinatif/cytologie , Activation des lymphocytes/immunologie , Antigènes CD27/immunologie , Transfert adoptif , Animaux , Antigènes viraux/immunologie , Antigène CD28/métabolisme , Centre germinatif/immunologie , Immunoglobulines/biosynthèse , Immunoglobulines/sang , Souris , Souris knockout , Infections à Orthomyxoviridae/immunologie , Rate/cytologie , Lymphocytes T/immunologie , Antigènes CD27/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE