Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(13)2023 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-37446306

RESUMEN

Fanconi anemia (FA) develops due to a mutation in one of the FANC genes that are involved in the repair of interstrand crosslinks (ICLs). FANCG, a member of the FA core complex, is essential for ICL repair. Previous FANCG-deficient mouse models were generated with drug-based selection cassettes in mixed mice backgrounds, leading to a disparity in the interpretation of genotype-related phenotype. We created a Fancg-KO (KO) mouse model using CRISPR/Cas9 to exclude these confounders. The entire Fancg locus was targeted and maintained on the immunological well-characterized C57BL/6J background. The intercrossing of heterozygous mice resulted in sub-Mendelian numbers of homozygous mice, suggesting the loss of FANCG can be embryonically lethal. KO mice displayed infertility and hypogonadism, but no other developmental problems. Bone marrow analysis revealed a defect in various hematopoietic stem and progenitor subsets with a bias towards myelopoiesis. Cell lines derived from Fancg-KO mice were hypersensitive to the crosslinking agents cisplatin and Mitomycin C, and Fancg-KO mouse embryonic fibroblasts (MEFs) displayed increased γ-H2AX upon cisplatin treatment. The reconstitution of these MEFs with Fancg cDNA corrected for the ICL hypersensitivity. This project provides a new, genetically, and immunologically well-defined Fancg-KO mouse model for further in vivo and in vitro studies on FANCG and ICL repair.


Asunto(s)
Cisplatino , Anemia de Fanconi , Humanos , Animales , Ratones , Cisplatino/metabolismo , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Ratones Endogámicos C57BL , Sistemas CRISPR-Cas , Proteínas de Unión al ADN/metabolismo , Fibroblastos/metabolismo , Mitomicina , Fenotipo , Proteína del Grupo de Complementación G de la Anemia de Fanconi/genética
2.
Proc Natl Acad Sci U S A ; 120(4): e2216055120, 2023 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-36669105

RESUMEN

DNA damage threatens genomic integrity and instigates stem cell failure. To bypass genotoxic lesions during replication, cells employ DNA damage tolerance (DDT), which is regulated via PCNA ubiquitination and REV1. DDT is conserved in all domains of life, yet its relevance in mammals remains unclear. Here, we show that inactivation of both PCNA-ubiquitination and REV1 results in embryonic and adult lethality, and the accumulation of DNA damage in hematopoietic stem and progenitor cells (HSPCs) that ultimately resulted in their depletion. Our results reveal the crucial relevance of DDT in the maintenance of stem cell compartments and mammalian life in unperturbed conditions.


Asunto(s)
Daño del ADN , Animales , Reparación del ADN , Replicación del ADN , Células Madre Hematopoyéticas/metabolismo , Mamíferos/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ubiquitinación
3.
Nucleic Acids Res ; 50(13): 7420-7435, 2022 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-35819193

RESUMEN

Crosslink repair depends on the Fanconi anemia pathway and translesion synthesis polymerases that replicate over unhooked crosslinks. Translesion synthesis is regulated via ubiquitination of PCNA, and independently via translesion synthesis polymerase REV1. The division of labor between PCNA-ubiquitination and REV1 in interstrand crosslink repair is unclear. Inhibition of either of these pathways has been proposed as a strategy to increase cytotoxicity of platinating agents in cancer treatment. Here, we defined the importance of PCNA-ubiquitination and REV1 for DNA in mammalian ICL repair. In mice, loss of PCNA-ubiquitination, but not REV1, resulted in germ cell defects and hypersensitivity to cisplatin. Loss of PCNA-ubiquitination, but not REV1 sensitized mammalian cancer cell lines to cisplatin. We identify polymerase Kappa as essential in tolerating DNA damage-induced lesions, in particular cisplatin lesions. Polk-deficient tumors were controlled by cisplatin treatment and it significantly delayed tumor outgrowth and increased overall survival of tumor bearing mice. Our results indicate that PCNA-ubiquitination and REV1 play distinct roles in DNA damage tolerance. Moreover, our results highlight POLK as a critical TLS polymerase in tolerating multiple genotoxic lesions, including cisplatin lesions. The relative frequent loss of Polk in cancers indicates an exploitable vulnerability for precision cancer medicine.


Asunto(s)
Reparación del ADN , Neoplasias , Animales , Cisplatino/uso terapéutico , Daño del ADN , Replicación del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Medicina de Precisión , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ubiquitinación
4.
Front Immunol ; 13: 986863, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36700204

RESUMEN

The development and differentiation of B cells is intimately linked to cell proliferation and the generation of diverse immunoglobulin gene (Ig) repertoires. The ubiquitin E3 ligase HUWE1 controls proliferation, DNA damage responses, and DNA repair, including the base excision repair (BER) pathway. These processes are of crucial importance for B-cell development in the bone marrow, and the germinal center (GC) response, which results in the clonal expansion and differentiation of B cells expressing high affinity immunoglobulins. Here, we re-examined the role of HUWE1 in B-cell proliferation and Ig gene diversification, focusing on its involvement in somatic hypermutation (SHM) and class switch recombination (CSR). B-cell-specific deletion of Huwe1 resulted in impaired development, differentiation and maturation of B cells in the bone marrow and peripheral lymphoid organs. HUWE1 deficiency diminished SHM and CSR by impairing B-cell proliferation and AID expression upon activation in vitro and in vivo, and was unrelated to the HUWE1-dependent regulation of the BER pathway. Interestingly, we found that HUWE1-deficient B cells showed increased mRNA expression of Myc target genes upon in vitro activation despite diminished proliferation. Our results confirm that the E3 ligase HUWE1 is an important contributor in coordinating the rapid transition of antigen naïve, resting B cells into antigen-activated B cells and regulates mutagenic processes in B cells by controlling AID expression and the post-transcriptional output of Myc target genes.


Asunto(s)
Cambio de Clase de Inmunoglobulina , Hipermutación Somática de Inmunoglobulina , Cambio de Clase de Inmunoglobulina/genética , Linfocitos B , Reparación del ADN , Diferenciación Celular/genética
5.
Front Pharmacol ; 12: 596535, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679394

RESUMEN

The antihelmintic drug ABZ and its metabolites belong to the chemical family of benzimidazoles (BZM) that act as potent tubulin polymerization inhibitors, suggesting a potential re-direction of BZMs for cancer therapy. Applying UV-Vis spectrometry we here demonstrate ABZ as a DNA intercalator. This insight led us to determine the primary mode of ABZ action in mammalian cells. As revealed by RNA sequencing, ABZ did neither grossly affect replication as analyzed by survival and replication stress signaling, nor the transcriptome. Actually, unbiased transcriptome analysis revealed a marked cell cycle signature in ABZ exposed cells. Indeed, short-term exposure to ABZ arrested mammalian cells in G2/M cell cycle stages associated with frequent gains and losses of chromatin. Cellular analyses revealed ABZ as a potent mammalian spindle poison for normal and malignant cells, explaining the serious chromosome segregation defects. Since chromosomal aberrations promote both cancer development and cell death, we determined if besides its general cytotoxicity, ABZ could predispose to tumor development. As measured by loss of heterozygosity (LOH) in vitro and in vivo ABZ was found as a potent inducer of LOH and accelerator of chromosomal missegregation.

6.
Proc Natl Acad Sci U S A ; 117(49): 31343-31352, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33229554

RESUMEN

Development of progenitor B cells (ProB cells) into precursor B cells (PreB cells) is dictated by immunoglobulin heavy chain checkpoint (IgHCC), where the IgHC encoded by a productively rearranged Igh allele assembles into a PreB cell receptor complex (PreBCR) to generate signals to initiate this transition and suppressing antigen receptor gene recombination, ensuring that only one productive Igh allele is expressed, a phenomenon known as Igh allelic exclusion. In contrast to a productively rearranged Igh allele, the Igh messenger RNA (mRNA) (IgHR) from a nonproductively rearranged Igh allele is degraded by nonsense-mediated decay (NMD). This fact prohibited firm conclusions regarding the contribution of stable IgHR to the molecular and developmental changes associated with the IgHCC. This point was addressed by generating the IghTer5H∆TM mouse model from IghTer5H mice having a premature termination codon at position +5 in leader exon of IghTer5H allele. This prohibited NMD, and the lack of a transmembrane region (∆TM) prevented the formation of any signaling-competent PreBCR complexes that may arise as a result of read-through translation across premature Ter5 stop codon. A highly sensitive sandwich Western blot revealed read-through translation of IghTer5H message, indicating that previous conclusions regarding a role of IgHR in establishing allelic exclusion requires further exploration. As determined by RNA sequencing (RNA-Seq), this low amount of IgHC sufficed to initiate PreB cell markers normally associated with PreBCR signaling. In contrast, the IghTer5H∆TM knock-in allele, which generated stable IgHR but no detectable IgHC, failed to induce PreB development. Our data indicate that the IgHCC is controlled at the level of IgHC and not IgHR expression.


Asunto(s)
Linfocitos B/citología , Linfocitos B/metabolismo , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/metabolismo , Alelos , Animales , Biomarcadores/metabolismo , Sitios Genéticos , Ratones Endogámicos C57BL , Células Precursoras de Linfocitos B/citología , Células Precursoras de Linfocitos B/inmunología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados
7.
Oncotarget ; 9(27): 18832-18843, 2018 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-29721165

RESUMEN

DNA damage tolerance (DDT) enables replication to continue in the presence of a damaged template and constitutes a key step in DNA interstrand crosslink repair. In this way DDT minimizes replication stress inflicted by a wide range of endogenous and exogenous agents, and provides a critical first line defense against alkylating and platinating chemotherapeutics. Effective DDT strongly depends on damage-induced, site-specific PCNA-ubiquitination at Lysine (K) 164 by the E2/E3 complex (RAD6/18). A survey of The Cancer Genome Atlas (TCGA) revealed a high frequency of tumors presents RAD6/RAD18 bi-allelic inactivating deletions. For instance, 11% of renal cell carcinoma and 5% of pancreatic tumors have inactivating RAD18-deletions and 7% of malignant peripheral nerve sheath tumors lack RAD6B. To determine the potential benefit for tumor-specific DDT defects, we followed a genetic approach by establishing unique sets of DDT-proficient PcnaK164 and -defective PcnaK164R lymphoma and breast cancer cell lines. In the absence of exogenous DNA damage, PcnaK164R tumors grew comparably to their PcnaK164 controls in vitro and in vivo. However, DDT-defective lymphomas and breast cancers were compared to their DDT-proficient controls hypersensitive to the chemotherapeutic drug cisplatin (CsPt), both in vitro and in vivo. CsPt strongly inhibited tumor growth and the overall survival of tumor bearing mice greatly improved in the DDT-defective condition. These insights open new therapeutic possibilities for precision cancer medicine with DNA damaging chemotherapeutics and optimize Next-Generation-Sequencing (NGS)-based cancer-diagnostics, -therapeutics, and -prognosis.

8.
Nucleic Acids Res ; 44(10): 4734-44, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-26926109

RESUMEN

PrimPol is a DNA damage tolerant polymerase displaying both translesion synthesis (TLS) and (re)-priming properties. This led us to study the consequences of a PrimPol deficiency in tolerating mutagenic lesions induced by members of the APOBEC/AID family of cytosine deaminases. Interestingly, during somatic hypermutation, PrimPol counteracts the generation of C>G transversions on the leading strand. Independently, mutation analyses in human invasive breast cancer confirmed a pro-mutagenic activity of APOBEC3B and revealed a genome-wide anti-mutagenic activity of PRIMPOL as well as most Y-family TLS polymerases. PRIMPOL especially prevents APOBEC3B targeted cytosine mutations within TpC dinucleotides. As C transversions induced by APOBEC/AID family members depend on the formation of AP-sites, we propose that PrimPol reprimes preferentially downstream of AP-sites on the leading strand, to prohibit error-prone TLS and simultaneously stimulate error-free homology directed repair. These in vivo studies are the first demonstrating a critical anti-mutagenic activity of PrimPol in genome maintenance.


Asunto(s)
Citidina Desaminasa/metabolismo , ADN Primasa/fisiología , ADN Polimerasa Dirigida por ADN/fisiología , Antígenos de Histocompatibilidad Menor/metabolismo , Enzimas Multifuncionales/fisiología , Mutagénesis , Animales , Linfocitos B/enzimología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Sistemas CRISPR-Cas , Línea Celular , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Citidina Desaminasa/antagonistas & inhibidores , ADN/metabolismo , Replicación del ADN , Femenino , Humanos , Cambio de Clase de Inmunoglobulina , Ratones Endogámicos C57BL , Hipermutación Somática de Inmunoglobulina , Linfocitos T/enzimología , Rayos Ultravioleta
9.
Data Brief ; 6: 556-61, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26909367

RESUMEN

The data described here provide genome-wide expression profiles of murine primitive hematopoietic stem and progenitor cells (LSK) and of B cell populations, obtained by high throughput sequencing. Cells are derived from wild-type mice and from mice deficient for the ubiquitin-specific protease 3 (USP3; Usp3Δ/Δ). Modification of histone proteins by ubiquitin plays a crucial role in the cellular response to DNA damage (DDR) (Jackson and Durocher, 2013) [1]. USP3 is a histone H2A deubiquitinating enzyme (DUB) that regulates ubiquitin-dependent DDR in response to DNA double-strand breaks (Nicassio et al., 2007; Doil et al., 2008) [2], [3]. Deletion of USP3 in mice increases the incidence of spontaneous tumors and affects hematopoiesis [4]. In particular, Usp3-knockout mice show progressive loss of B and T cells and decreased functional potential of hematopoietic stem cells (HSCs) during aging. USP3-deficient cells, including HSCs, display enhanced histone ubiquitination, accumulate spontaneous DNA damage and are hypersensitive to ionizing radiation (Lancini et al., 2014) [4]. To address whether USP3 loss leads to deregulation of specific molecular pathways relevant to HSC homeostasis and/or B cell development, we have employed the RNA-sequencing technology and investigated transcriptional differences between wild-type and Usp3Δ/Δ LSK, naïve B cells or in vitro activated B cells. The data relate to the research article "Tight regulation of ubiquitin-mediated DNA damage response by USP3 preserves the functional integrity of hematopoietic stem cells" (Lancini et al., 2014) [4]. The RNA-sequencing and analysis data sets have been deposited in NCBI׳s Gene Expression Omnibus (Edgar et al., 2002) [5] and are accessible through GEO Series accession number GSE58495 (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE58495). With this article, we present validation of the RNA-seq data set through quantitative real-time PCR and comparative analysis.

10.
Nucleic Acids Res ; 43(1): 282-94, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25505145

RESUMEN

Translesion synthesis (TLS) provides a highly conserved mechanism that enables DNA synthesis on a damaged template. TLS is performed by specialized DNA polymerases of which polymerase (Pol) κ is important for the cellular response to DNA damage induced by benzo[a]pyrene-7,8-dihydrodiol-9,10-epoxide (BPDE), ultraviolet (UV) light and the alkylating agent methyl methanesulfonate (MMS). As TLS polymerases are intrinsically error-prone, tight regulation of their activity is required. One level of control is provided by ubiquitination of the homotrimeric DNA clamp PCNA at lysine residue 164 (PCNA-Ub). We here show that Polκ can function independently of PCNA modification and that Polη can function as a backup during TLS of MMS-induced lesions. Compared to cell lines deficient for PCNA modification (Pcna(K164R)) or Polκ, double mutant cell lines display hypersensitivity to MMS but not to BPDE or UV-C. Double mutant cells also displayed delayed post-replicative TLS, accumulate higher levels of replication stress and delayed S-phase progression. Furthermore, we show that Polη and Polκ are redundant in the DNA damage bypass of MMS-induced DNA damage. Taken together, we provide evidence for PCNA-Ub-independent activation of Polκ and establish Polη as an important backup polymerase in the absence of Polκ in response to MMS-induced DNA damage.


Asunto(s)
Daño del ADN , ADN Polimerasa Dirigida por ADN/fisiología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ubiquitinación , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Supervivencia Celular , Células Cultivadas , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Replicación del ADN , ADN Polimerasa Dirigida por ADN/genética , Metilmetanosulfonato/toxicidad , Ratones Noqueados , Mutación , Antígeno Nuclear de Célula en Proliferación/genética , Proteínas Quinasas/metabolismo , Fase S
11.
J Exp Med ; 211(9): 1759-77, 2014 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-25113974

RESUMEN

Histone ubiquitination at DNA breaks is required for activation of the DNA damage response (DDR) and DNA repair. How the dynamic removal of this modification by deubiquitinating enzymes (DUBs) impacts genome maintenance in vivo is largely unknown. To address this question, we generated mice deficient for Ub-specific protease 3 (USP3; Usp3Δ/Δ), a histone H2A DUB which negatively regulates ubiquitin-dependent DDR signaling. Notably, USP3 deletion increased the levels of histone ubiquitination in adult tissues, reduced the hematopoietic stem cell (HSC) reserves over time, and shortened animal life span. Mechanistically, our data show that USP3 is important in HSC homeostasis, preserving HSC self-renewal, and repopulation potential in vivo and proliferation in vitro. A defective DDR and unresolved spontaneous DNA damage contribute to cell cycle restriction of Usp3Δ/Δ HSCs. Beyond the hematopoietic system, Usp3Δ/Δ animals spontaneously developed tumors, and primary Usp3Δ/Δ cells failed to preserve chromosomal integrity. These findings broadly support the regulation of chromatin ubiquitination as a key pathway in preserving tissue function through modulation of the response to genotoxic stress.


Asunto(s)
Daño del ADN/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Animales , Carcinogénesis , Proliferación Celular , Senescencia Celular , Roturas del ADN de Doble Cadena , Reparación del ADN/fisiología , Femenino , Histonas/metabolismo , Homeostasis , Linfopenia/etiología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteasas Ubiquitina-Específicas/deficiencia , Proteasas Ubiquitina-Específicas/genética , Ubiquitinación
12.
PLoS One ; 8(7): e69815, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23922811

RESUMEN

The Aicda locus encodes the activation induced cytidine deaminase (AID) and is highly expressed in germinal center (GC) B cells to initiate somatic hypermutation (SHM) and class switch recombination (CSR) of immunoglobulin (Ig) genes. Besides these Ig specific activities in B cells, AID has been implicated in active DNA demethylation in non-B cell systems. We here determined a potential role of AID as an epigenetic eraser and transcriptional regulator in B cells. RNA-Seq on different B cell subsets revealed that Aicda(-/-) B cells are developmentally affected. However as shown by RNA-Seq, MethylCap-Seq, and SNP analysis these transcriptome alterations may not relate to AID, but alternatively to a CBA mouse strain derived region around the targeted Aicda locus. These unexpected confounding parameters provide alternative, AID-independent interpretations on genotype-phenotype correlations previously reported in numerous studies on AID using the Aicda(-/-) mouse strain.


Asunto(s)
Linfocitos B/enzimología , Linfocitos B/metabolismo , Citidina Desaminasa/deficiencia , Animales , Citidina Desaminasa/genética , Metilación de ADN/genética , Genotipo , Ratones , Ratones Mutantes
13.
DNA Repair (Amst) ; 10(10): 1051-9, 2011 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-21889916

RESUMEN

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.


Asunto(s)
Linfocitos B/metabolismo , Daño del ADN/genética , Reparación del ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Hipermutación Somática de Inmunoglobulina/genética , Ubiquitinación , Animales , Linfocitos B/citología , Activación Enzimática , Lisina/genética , Ratones , Ratones Endogámicos C57BL , Mutagénesis , Mutación , Antígeno Nuclear de Célula en Proliferación/genética , Rayos Ultravioleta
14.
PLoS One ; 6(1): e16669, 2011 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-21304913

RESUMEN

Monoubiquitylation of the homotrimeric DNA sliding clamp PCNA at lysine residue 164 (PCNA(K164)) is a highly conserved, DNA damage-inducible process that is mediated by the E2/E3 complex Rad6/Rad18. This ubiquitylation event recruits translesion synthesis (TLS) polymerases capable of replicating across damaged DNA templates. Besides PCNA, the Rad6/Rad18 complex was recently shown in yeast to ubiquitylate also 9-1-1, a heterotrimeric DNA sliding clamp composed of Rad9, Rad1, and Hus1 in a DNA damage-inducible manner. Based on the highly similar crystal structures of PCNA and 9-1-1, K185 of Rad1 (Rad1(K185)) was identified as the only topological equivalent of PCNA(K164). To investigate a potential role of posttranslational modifications of Rad1(K185) in DNA damage management, we here generated a mouse model with a conditional deletable Rad1(K185R) allele. The Rad1(K185) residue was found to be dispensable for Chk1 activation, DNA damage survival, and class switch recombination of immunoglobulin genes as well as recruitment of TLS polymerases during somatic hypermutation of immunoglobulin genes. Our data indicate that Rad1(K185) is not a functional counterpart of PCNA(K164).


Asunto(s)
Exonucleasas/fisiología , Lisina/fisiología , Antígeno Nuclear de Célula en Proliferación/fisiología , Animales , Daño del ADN , Ratones , Modelos Animales , Procesamiento Proteico-Postraduccional
15.
DNA Repair (Amst) ; 10(4): 438-44, 2011 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-21269891

RESUMEN

DNA damage tolerance is regulated at least in part at the level of proliferating cell nuclear antigen (PCNA) ubiquitination. Monoubiquitination (PCNA-Ub) at lysine residue 164 (K164) stimulates error-prone translesion synthesis (TLS), Rad5-dependent polyubiquitination (PCNA-Ub(n)) stimulates error-free template switching (TS). To generate high affinity antibodies by somatic hypermutation (SHM), B cells profit from error-prone TLS polymerases. Consistent with the role of PCNA-Ub in stimulating TLS, hypermutated B cells of PCNA(K164R) mutant mice display a defect in generating selective point mutations. Two Rad5 orthologs, HLTF and SHPRH have been identified as alternative E3 ligases generating PCNA-Ub(n) in mammals. As PCNA-Ub and PCNA-Ub(n) both make use of K164, error-free PCNA-Ub(n)-dependent TS may suppress error-prone PCNA-Ub-dependent TLS. To determine a regulatory role of Shprh and Hltf in SHM, we generated Shprh/Hltf double mutant mice. Interestingly, while the formation of PCNA-Ub and PCNA-Ub(n) is prohibited in PCNA(K164R) MEFs, the formation of PCNA-Ub(n) is not abolished in Shprh/Hltf mutant MEFs. In line with these observations Shprh/Hltf double mutant B cells were not hypersensitive to DNA damage. Furthermore, SHM was normal in Shprh/Hltf mutant B cells. These data suggest the existence of an alternative E3 ligase in the generation of PCNA-Ub(n).


Asunto(s)
ADN Helicasas/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Hipermutación Somática de Inmunoglobulina/genética , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/fisiología , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Reactivos de Enlaces Cruzados/farmacología , ADN Helicasas/deficiencia , ADN Helicasas/genética , Células Madre Embrionarias/metabolismo , Técnicas de Silenciamiento del Gen , Orden Génico , Marcación de Gen , Cambio de Clase de Inmunoglobulina/efectos de los fármacos , Lipopolisacáridos/farmacología , Ratones , Ratones Noqueados , Antígeno Nuclear de Célula en Proliferación/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Ubiquitinación/genética , Ubiquitinación/efectos de la radiación , Rayos Ultravioleta/efectos adversos
16.
PLoS One ; 5(12): e15236, 2010 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-21209924

RESUMEN

To generate high affinity antibodies during an immune response, B cells undergo somatic hypermutation (SHM) of their immunoglobulin genes. Error-prone translesion synthesis (TLS) DNA polymerases have been reported to be responsible for all mutations at template A/T and at least a fraction of G/C transversions. In contrast to A/T mutations which depend on PCNA ubiquitination, it remains unclear how G/C transversions are regulated during SHM. Several lines of evidence indicate a mechanistic link between the Fanconi Anemia (FA) pathway and TLS. To investigate the contribution of the FA pathway in SHM we analyzed FancG-deficient B cells. B cells deficient for FancG, an essential member of the FA core complex, were hypersensitive to treatment with cross-linking agents. However, the frequencies and nucleotide exchange spectra of SHM remained comparable between wild-type and FancG-deficient B cells. These data indicate that the FA pathway is not involved in regulating the outcome of SHM in mammals. In addition, the FA pathway appears dispensable for class switch recombination.


Asunto(s)
Anemia de Fanconi/metabolismo , Mutación , Animales , Linfocitos B/citología , Cisplatino/farmacología , Reactivos de Enlaces Cruzados/farmacología , ADN/química , Análisis Mutacional de ADN , Fibroblastos/citología , Genotipo , Cambio de Clase de Inmunoglobulina , Ratones , Fenotipo , Ubiquitina/química , Rayos Ultravioleta
17.
J Exp Med ; 206(12): 2603-11, 2009 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-19901081

RESUMEN

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.


Asunto(s)
Linfocitos B/metabolismo , Proteína 2 Homóloga a MutS/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Hipermutación Somática de Inmunoglobulina/fisiología , Uracil-ADN Glicosidasa/metabolismo , Animales , Afinidad de Anticuerpos/fisiología , Linfocitos B/inmunología , Ratones , Ratones Mutantes , Proteína 2 Homóloga a MutS/genética , Proteína 2 Homóloga a MutS/inmunología , Mutación/inmunología , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/inmunología , Proteínas Ubiquitinadas/genética , Proteínas Ubiquitinadas/inmunología , Proteínas Ubiquitinadas/metabolismo , Uracil-ADN Glicosidasa/genética , Uracil-ADN Glicosidasa/inmunología
18.
Philos Trans R Soc Lond B Biol Sci ; 364(1517): 621-9, 2009 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-19008189

RESUMEN

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.


Asunto(s)
Reparación del ADN , Antígeno Nuclear de Célula en Proliferación/genética , Hipermutación Somática de Inmunoglobulina/genética , Animales , Análisis Mutacional de ADN , ADN Polimerasa Dirigida por ADN/metabolismo , Ratones , Ratones Mutantes , Modelos Genéticos , Nucleotidiltransferasas/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ubiquitinación
19.
J Exp Med ; 204(8): 1989-98, 2007 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-17664295

RESUMEN

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.


Asunto(s)
Adenina/química , Citosina/química , Regulación de la Expresión Génica , Inmunoglobulinas/genética , Mutagénesis , Mutación , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/fisiología , Animales , Linfocitos B/metabolismo , Proliferación Celular , ADN Polimerasa Dirigida por ADN/metabolismo , Homocigoto , Ratones , Modelos Biológicos , Fenotipo , Ubiquitina/química , Ubiquitina/metabolismo
20.
J Clin Oncol ; 23(35): 8978-91, 2005 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-16260696

RESUMEN

PURPOSE: To determine the feasibility, toxicity, and immunologic effects of vaccination with autologous tumor cells retrovirally transduced with the GM-CSF gene, we performed a phase I/II vaccination study in stage IV metastatic melanoma patients. PATIENTS AND METHODS: Sixty-four patients were randomly assigned to receive three vaccinations of high-dose or low-dose tumor cells at 3-week intervals. Tumor cell vaccine preparation succeeded for 56 patients (88%), but because of progressive disease, the well-tolerated vaccination was completed in only 28 patients. We analyzed the priming of T cells against melanoma antigens, MART-1, tyrosinase, gp100, MAGE-A1, and MAGE-A3 using human leukocyte antigen/peptide tetramers and functional assays. RESULTS: The high-dose vaccination induced the infiltration of T cells into the tumor tissue. Three of 14 patients receiving the high-dose vaccine showed an increase in MART-1- or gp100-specific T cells in the peripheral blood during vaccination. Six patients experienced disease-free survival for more than 5 years, and two of these patients developed vitiligo at multiple sites after vaccination. MART-1- and gp100-specific T cells were found infiltrating in vitiligo skin. Upon vaccination, the T cells acquired an effector phenotype and produced interferon-gamma on specific antigenic stimulation. CONCLUSION: We conclude that vaccination with GM-CSF-transduced autologous tumor cells has limited toxicity and can enhance T-cell activation against melanocyte differentiation antigens, which can lead to vitiligo. Whether the induction of autoimmune vitiligo may prolong disease-free survival of metastatic melanoma patients who are surgically rendered as having no evidence of disease before vaccination is worthy of further investigation.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Melanoma/tratamiento farmacológico , Adyuvantes Inmunológicos , Adulto , Anciano , Antígenos de Neoplasias/inmunología , Enfermedades Autoinmunes/etiología , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/inmunología , Citotoxicidad Inmunológica , Esquema de Medicación , Estudios de Factibilidad , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/efectos adversos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Masculino , Melanoma/inmunología , Persona de Mediana Edad , Proteínas de Neoplasias/inmunología , Fragmentos de Péptidos/inmunología , Linfocitos T/inmunología , Vitíligo/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...