Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Cell ; 153(6): 1266-80, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23727112

RESUMEN

The DNA damage response (DDR) protein 53BP1 protects DNA ends from excessive resection in G1, and thereby favors repair by nonhomologous end-joining (NHEJ) as opposed to homologous recombination (HR). During S phase, BRCA1 antagonizes 53BP1 to promote HR. The pro-NHEJ and antirecombinase functions of 53BP1 are mediated in part by RIF1, the only known factor that requires 53BP1 phosphorylation for its recruitment to double-strand breaks (DSBs). Here, we show that a 53BP1 phosphomutant, 53BP18A, comprising alanine substitutions of the eight most N-terminal S/TQ phosphorylation sites, mimics 53BP1 deficiency by restoring genome stability in BRCA1-deficient cells yet behaves like wild-type 53BP1 with respect to immunoglobulin class switch recombination (CSR). 53BP18A recruits RIF1 but fails to recruit the DDR protein PTIP to DSBs, and disruption of PTIP phenocopies 53BP18A. We conclude that 53BP1 promotes productive CSR and suppresses mutagenic DNA repair through distinct phosphodependent interactions with RIF1 and PTIP.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/metabolismo , Cambio de Clase de Inmunoglobulina , Proteínas Nucleares/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Animales , Linfocitos B/metabolismo , Proteína BRCA1/metabolismo , Proteínas Cromosómicas no Histona/genética , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/metabolismo , Inestabilidad Genómica , Ratones , Mutación , Proteína 1 de Unión al Supresor Tumoral P53
2.
Trends Immunol ; 44(10): 782-791, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37640588

RESUMEN

The DNA double-strand break (DSB) repair factor 53BP1 has long been implicated in V(D)J and class switch recombination (CSR) of mammalian lymphocyte receptors. However, the dissection of the underlying molecular activities is hampered by a paucity of studies [V(D)J] and plurality of phenotypes (CSR) associated with 53BP1 deficiency. Here, we revisit the currently accepted roles of 53BP1 in antibody diversification in view of the recent identification of its downstream effectors in DSB protection and latest advances in genome architecture. We propose that, in addition to end protection, 53BP1-mediated end-tethering stabilization is essential for CSR. Furthermore, we support a pre-DSB role during V(D)J recombination. Our perspective underscores the importance of evaluating repair of DSBs in relation to their dynamic architectural contexts.


Asunto(s)
Anticuerpos , Roturas del ADN de Doble Cadena , Reparación del ADN , Proteína 1 de Unión al Supresor Tumoral P53 , Animales , Humanos , Ratones , Anticuerpos/genética , Cambio de Clase de Inmunoglobulina/genética , Linfocitos , Mamíferos
3.
Cell ; 147(1): 95-106, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21962510

RESUMEN

Chromosomal rearrangements, including translocations, require formation and joining of DNA double strand breaks (DSBs). These events disrupt the integrity of the genome and are frequently involved in producing leukemias, lymphomas and sarcomas. Despite the importance of these events, current understanding of their genesis is limited. To examine the origins of chromosomal rearrangements we developed Translocation Capture Sequencing (TC-Seq), a method to document chromosomal rearrangements genome-wide, in primary cells. We examined over 180,000 rearrangements obtained from 400 million B lymphocytes, revealing that proximity between DSBs, transcriptional activity and chromosome territories are key determinants of genome rearrangement. Specifically, rearrangements tend to occur in cis and to transcribed genes. Finally, we find that activation-induced cytidine deaminase (AID) induces the rearrangement of many genes found as translocation partners in mature B cell lymphoma.


Asunto(s)
Linfocitos B/metabolismo , Genoma , Mutagénesis , Translocación Genética , Animales , Células Cultivadas , Citidina Desaminasa/metabolismo , Genes myc , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Ratones , Neoplasias/genética , Análisis de Secuencia de ADN/métodos , Bazo/citología
4.
Mol Cell ; 72(4): 636-649.e8, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30293785

RESUMEN

Class switch recombination (CSR) is a DNA recombination reaction that diversifies the effector component of antibody responses. CSR is initiated by activation-induced cytidine deaminase (AID), which targets transcriptionally active immunoglobulin heavy chain (Igh) switch donor and acceptor DNA. The 3' Igh super-enhancer, 3' regulatory region (3'RR), is essential for acceptor region transcription, but how this function is regulated is unknown. Here, we identify the chromatin reader ZMYND8 as an essential regulator of the 3'RR. In B cells, ZMYND8 binds promoters and super-enhancers, including the Igh enhancers. ZMYND8 controls the 3'RR activity by modulating the enhancer transcriptional status. In its absence, there is increased 3'RR polymerase loading and decreased acceptor region transcription and CSR. In addition to CSR, ZMYND8 deficiency impairs somatic hypermutation (SHM) of Igh, which is also dependent on the 3'RR. Thus, ZMYND8 controls Igh diversification in mature B lymphocytes by regulating the activity of the 3' Igh super-enhancer.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Cambio de Clase de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/genética , Proteínas Supresoras de Tumor/genética , Animales , Linfocitos B , Línea Celular , Cromatina/genética , Cromatina/metabolismo , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , ADN/genética , Elementos de Facilitación Genéticos , Reordenamiento Génico , Humanos , Dominios MYND , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Secuencias Reguladoras de Ácidos Nucleicos , Hipermutación Somática de Inmunoglobulina/genética , Proteínas Supresoras de Tumor/metabolismo
5.
Trends Biochem Sci ; 46(3): 184-199, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33250286

RESUMEN

Immunoglobulin (Ig) class switch recombination (CSR) is the process occurring in mature B cells that diversifies the effector component of antibody responses. CSR is initiated by the activity of the B cell-specific enzyme activation-induced cytidine deaminase (AID), which leads to the formation of programmed DNA double-strand breaks (DSBs) at the Ig heavy chain (Igh) locus. Mature B cells use a multilayered and complex regulatory framework to ensure that AID-induced DNA breaks are channeled into productive repair reactions leading to CSR, and to avoid aberrant repair events causing lymphomagenic chromosomal translocations. Here, we review the DNA repair pathways acting on AID-induced DSBs and their functional interplay, with a particular focus on the latest developments in their molecular composition and mechanistic regulation.


Asunto(s)
Roturas del ADN de Doble Cadena , Cambio de Clase de Inmunoglobulina , Linfocitos B , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Reparación del ADN , Cadenas Pesadas de Inmunoglobulina/genética
6.
Cell ; 143(1): 122-33, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20887897

RESUMEN

Activation-induced cytidine deaminase (AID) initiates antibody gene diversification by creating U:G mismatches. However, AID is not specific for antibody genes; Off-target lesions can activate oncogenes or cause chromosome translocations. Despite its importance in these transactions little is known about how AID finds its targets. We performed an shRNA screen to identify factors required for class switch recombination (CSR) of antibody loci. We found that Spt5, a factor associated with stalled RNA polymerase II (Pol II) and single stranded DNA (ssDNA), is required for CSR. Spt5 interacts with AID, it facilitates association between AID and Pol II, and AID recruitment to its Ig and non-Ig targets. ChIP-seq experiments reveal that Spt5 colocalizes with AID and stalled Pol II. Further, Spt5 accumulation at sites of Pol II stalling is predictive of AID-induced mutation. We propose that AID is targeted to sites of Pol II stalling in part via its association with Spt5.


Asunto(s)
Linfocitos B/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Citidina Desaminasa/metabolismo , Cambio de Clase de Inmunoglobulina , ARN Polimerasa II/metabolismo , Factores de Elongación Transcripcional/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Fibroblastos/metabolismo , Humanos , Inmunoglobulinas/genética , Ratones
7.
Immunity ; 43(6): 1075-86, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26620760

RESUMEN

Phosphatidylinositol 3' OH kinase (PI3K) signaling and FOXO transcription factors play opposing roles at several B cell developmental stages. We show here abundant nuclear FOXO1 expression in the proliferative compartment of the germinal center (GC), its dark zone (DZ), and PI3K activity, downregulating FOXO1, in the light zone (LZ), where cells are selected for further differentiation. In the LZ, however, FOXO1 was expressed in a fraction of cells destined for DZ reentry. Upon FOXO1 ablation or induction of PI3K activity, GCs lost their DZ, owing at least partly to downregulation of the chemokine receptor CXCR4. Although this prevented proper cyclic selection of cells in GCs, somatic hypermutation and proliferation were maintained. Class switch recombination was partly lost due to a failure of switch region targeting by activation-induced deaminase (AID).


Asunto(s)
Linfocitos B/inmunología , Diferenciación Celular/inmunología , Factores de Transcripción Forkhead/inmunología , Centro Germinal/inmunología , Fosfatidilinositol 3-Quinasas/inmunología , Animales , Linfocitos B/citología , Separación Celular , Cromatografía Liquida , Citidina Desaminasa/inmunología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteína Forkhead Box O1 , Regulación de la Expresión Génica/inmunología , Centro Germinal/citología , Cambio de Clase de Inmunoglobulina/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Mutantes , Reacción en Cadena de la Polimerasa , Hipermutación Somática de Inmunoglobulina/inmunología , Espectrometría de Masas en Tándem
8.
Int J Mol Sci ; 24(3)2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36769095

RESUMEN

Critical illness myopathy (CIM) is an acquired, devastating, multifactorial muscle-wasting disease with incomplete recovery. The impact on hospital costs and permanent loss of quality of life is enormous. Incomplete recovery might imply that the function of muscle stem cells (MuSC) is impaired. We tested whether epigenetic alterations could be in part responsible. We characterized human muscle stem cells (MuSC) isolated from early CIM and analyzed epigenetic alterations (CIM n = 15, controls n = 21) by RNA-Seq, immunofluorescence, analysis of DNA repair, and ATAC-Seq. CIM-MuSC were transplanted into immunodeficient NOG mice to assess their regenerative potential. CIM-MuSC exhibited significant growth deficits, reduced ability to differentiate into myotubes, and impaired DNA repair. The chromatin structure was damaged, as characterized by alterations in mRNA of histone 1, depletion or dislocation of core proteins of nucleosome remodeling and deacetylase complex, and loosening of multiple nucleosome-spanning sites. Functionally, CIM-MuSC had a defect in building new muscle fibers. Further, MuSC obtained from the electrically stimulated muscle of CIM patients was very similar to control MuSC, indicating the impact of muscle contraction in the onset of CIM. CIM not only affects working skeletal muscle but has a lasting and severe epigenetic impact on MuSC.


Asunto(s)
Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2 , Enfermedades Musculares , Humanos , Animales , Ratones , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Enfermedad Crítica , Calidad de Vida , Enfermedades Musculares/metabolismo , Músculo Esquelético/metabolismo , Células Madre
10.
Blood ; 131(12): 1311-1324, 2018 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-29348130

RESUMEN

The regenerative capacity of hematopoietic stem cells (HSCs) is limited by the accumulation of DNA damage. Conditional mutagenesis of the histone 3 lysine 4 (H3K4) methyltransferase, Setd1a, revealed that it is required for the expression of DNA damage recognition and repair pathways in HSCs. Specific deletion of Setd1a in adult long-term (LT) HSCs is compatible with adult life and has little effect on the maintenance of phenotypic LT-HSCs in the bone marrow. However, SETD1A-deficient LT-HSCs lose their transcriptional cellular identity, accompanied by loss of their proliferative capacity and stem cell function under replicative stress in situ and after transplantation. In response to inflammatory stimulation, SETD1A protects HSCs and progenitors from activation-induced attrition in vivo. The comprehensive regulation of DNA damage responses by SETD1A in HSCs is clearly distinct from the key roles played by other epigenetic regulators, including the major leukemogenic H3K4 methyltransferase MLL1, or MLL5, indicating that HSC identity and function is supported by cooperative specificities within an epigenetic framework.


Asunto(s)
Proliferación Celular , Daño del ADN , Reparación del ADN , Células Madre Hematopoyéticas/enzimología , N-Metiltransferasa de Histona-Lisina/metabolismo , Animales , N-Metiltransferasa de Histona-Lisina/genética , Ratones , Ratones Noqueados , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo
11.
Mol Cell ; 42(3): 319-29, 2011 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-21549309

RESUMEN

53BP1 is a DNA damage protein that forms phosphorylated H2AX (γ-H2AX) dependent foci in a 1 Mb region surrounding DNA double-strand breaks (DSBs). In addition, 53BP1 promotes genomic stability by regulating the metabolism of DNA ends. We have compared the joining rates of paired DSBs separated by 1.2 kb to 27 Mb on chromosome 12 in the presence or absence of 53BP1. 53BP1 facilitates joining of intrachromosomal DSBs but only at distances corresponding to γ-H2AX spreading. In contrast, DNA end protection by 53BP1 is distance independent. Furthermore, analysis of 53BP1 mutants shows that chromatin association, oligomerization, and N-terminal ATM phosphorylation are all required for DNA end protection and joining as measured by immunoglobulin class switch recombination. These data elucidate the molecular events that are required for 53BP1 to maintain genomic stability and point to a model wherein 53BP1 and H2AX cooperate to repress resection of DSBs.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/genética , Cambio de Clase de Inmunoglobulina/genética , Animales , Linfocitos B/metabolismo , Sitios de Unión , Western Blotting , Células Cultivadas , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Citometría de Flujo , Histonas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Genéticos , Fosforilación , Multimerización de Proteína , Recombinación Genética , Proteína 1 de Unión al Supresor Tumoral P53
12.
Hum Mol Genet ; 24(25): 7361-72, 2015 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-26476407

RESUMEN

Null mutations in genes involved in V(D)J recombination cause a block in B- and T-cell development, clinically presenting as severe combined immunodeficiency (SCID). Hypomorphic mutations in the non-homologous end-joining gene DCLRE1C (encoding ARTEMIS) have been described to cause atypical SCID, Omenn syndrome, Hyper IgM syndrome and inflammatory bowel disease-all with severely impaired T-cell immunity. By whole-exome sequencing, we investigated the molecular defect in a consanguineous family with three children clinically diagnosed with antibody deficiency. We identified perfectly segregating homozygous variants in DCLRE1C in three index patients with recurrent respiratory tract infections, very low B-cell numbers and serum IgA levels. In patients, decreased colony survival after irradiation, impaired proliferative response and reduced counts of naïve T cells were observed in addition to a restricted T-cell receptor repertoire, increased palindromic nucleotides in the complementarity determining regions 3 and long stretches of microhomology at switch junctions. Defective V(D)J recombination was complemented by wild-type ARTEMIS protein in vitro. Subsequently, homozygous or compound heterozygous DCLRE1C mutations were identified in nine patients from the same geographic region. We demonstrate that DCLRE1C mutations can cause a phenotype presenting as only antibody deficiency. This novel association broadens the clinical spectrum associated with ARTEMIS mutations. Clinicians should consider the possibility that an immunodeficiency with a clinically mild initial presentation could be a combined immunodeficiency, so as to provide appropriate care for affected patients.


Asunto(s)
Proteínas Nucleares/genética , Inmunodeficiencia Combinada Grave/genética , Linfocitos B/metabolismo , Niño , Preescolar , Proteínas de Unión al ADN , Endonucleasas , Femenino , Humanos , Inmunoglobulina A/metabolismo , Masculino , Mutación/genética
13.
Immunity ; 28(5): 630-8, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18455451

RESUMEN

MicroRNAs (miRNAs) are small noncoding RNAs that regulate vast networks of genes that share miRNA target sequences. To examine the physiologic effects of an individual miRNA-mRNA interaction in vivo, we generated mice that carry a mutation in the putative microRNA-155 (miR-155) binding site in the 3'-untranslated region of activation-induced cytidine deaminase (AID), designated Aicda(155) mice. AID is required for immunoglobulin gene diversification in B lymphocytes, but it also promotes chromosomal translocations. Aicda(155) caused an increase in steady-state Aicda mRNA and protein amounts by increasing the half-life of the mRNA, resulting in a high degree of Myc-Igh translocations. A similar but more pronounced translocation phenotype was also found in miR-155-deficient mice. Our experiments indicate that miR-155 can act as a tumor suppressor by reducing potentially oncogenic translocations generated by AID.


Asunto(s)
Linfocitos B/enzimología , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Genes myc , Cadenas Pesadas de Inmunoglobulina/genética , MicroARNs/metabolismo , Translocación Genética , Regiones no Traducidas 3' , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Genes de Inmunoglobulinas , Cambio de Clase de Inmunoglobulina , Lipopolisacáridos/inmunología , Ratones , Ratones Mutantes , MicroARNs/genética , Mutación , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Hipermutación Somática de Inmunoglobulina
14.
Mol Cell ; 34(3): 285-97, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19450527

RESUMEN

The DNA double-strand break (DSB) repair protein DNA-PKcs and the signal transducer ATM are both activated by DNA breaks and phosphorylate similar substrates in vitro, yet appear to have distinct functions in vivo. Here, we show that ATM and DNA-PKcs have overlapping functions in lymphocytes. Ablation of both kinase activities in cells undergoing immunoglobulin class switch recombination leads to a compound defect in switching and a synergistic increase in chromosomal fragmentation, DNA insertions, and translocations due to aberrant processing of DSBs. These abnormalities are attributed to a compound deficiency in phosphorylation of key proteins required for DNA repair, class switching, and cell death. Notably, both kinases are required for normal levels of p53 phosphorylation in B and T cells and p53-dependent apoptosis. Our experiments reveal a DNA-PKcs-dependent pathway that regulates DNA repair and activation of p53 in the absence of ATM.


Asunto(s)
Apoptosis/fisiología , Proteínas de Ciclo Celular/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Linfocitos/fisiología , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Secuencia de Bases , Proteínas de Ciclo Celular/genética , Células Cultivadas , Proteína Quinasa Activada por ADN/genética , Proteínas de Unión al ADN/genética , Fibroblastos/citología , Fibroblastos/fisiología , Inestabilidad Genómica , Cambio de Clase de Inmunoglobulina , Linfocitos/citología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Timo/citología , Proteína 28 que Contiene Motivos Tripartito , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética
15.
J Allergy Clin Immunol ; 135(4): 998-1007.e6, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25312759

RESUMEN

BACKGROUND: Immunoglobulin class-switch recombination defects (CSR-D) are rare primary immunodeficiencies characterized by impaired production of switched immunoglobulin isotypes and normal or elevated IgM levels. They are caused by impaired T:B cooperation or intrinsic B cell defects. However, many immunoglobulin CSR-Ds are still undefined at the molecular level. OBJECTIVE: This study's objective was to delineate new causes of immunoglobulin CSR-Ds and thus gain further insights into the process of immunoglobulin class-switch recombination (CSR). METHODS: Exome sequencing in 2 immunoglobulin CSR-D patients identified variations in the INO80 gene. Functional experiments were performed to assess the function of INO80 on immunoglobulin CSR. RESULTS: We identified recessive, nonsynonymous coding variations in the INO80 gene in 2 patients affected by defective immunoglobulin CSR. Expression of wild-type INO80 in patients' fibroblastic cells corrected their hypersensitivity to high doses of γ-irradiation. In murine CH12-F3 cells, the INO80 complex accumulates at Sα and Eµ regions of the IgH locus, and downregulation of INO80 as well as its partners Reptin and Pontin impaired CSR. In addition, Reptin and Pontin were shown to interact with activation-induced cytidine deaminase. Finally, an abnormal separation of sister chromatids was observed upon INO80 downregulation in CH12-F3 cells, pinpointing its role in cohesin activity. CONCLUSION: INO80 deficiency appears to be associated with defective immunoglobulin CSR. We propose that the INO80 complex modulates cohesin function that may be required during immunoglobulin switch region synapsis.


Asunto(s)
Ensamble y Desensamble de Cromatina , ADN Helicasas/genética , Reordenamiento Génico , Cambio de Clase de Inmunoglobulina , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , ATPasas Asociadas con Actividades Celulares Diversas , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Supervivencia Celular/genética , Proteínas Cromosómicas no Histona/metabolismo , ADN Helicasas/metabolismo , Reparación del ADN , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Variación Genética , Humanos , Isotipos de Inmunoglobulinas/genética , Región de Cambio de la Inmunoglobulina , Síndromes de Inmunodeficiencia/metabolismo , Modelos Biológicos , Unión Proteica , Transporte de Proteínas , Cohesinas
16.
Nat Commun ; 14(1): 8049, 2023 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-38081811

RESUMEN

The mammalian DNA replication timing (RT) program is crucial for the proper functioning and integrity of the genome. The best-known mechanism for controlling RT is the suppression of late origins of replication in heterochromatin by RIF1. Here, we report that in antigen-activated, hypermutating murine B lymphocytes, RIF1 binds predominantly to early-replicating active chromatin and promotes early replication, but plays a minor role in regulating replication origin activity, gene expression and genome organization in B cells. Furthermore, we find that RIF1 functions in a complementary and non-epistatic manner with minichromosome maintenance (MCM) proteins to establish early RT signatures genome-wide and, specifically, to ensure the early replication of highly transcribed genes. These findings reveal additional layers of regulation within the B cell RT program, driven by the coordinated activity of RIF1 and MCM proteins.


Asunto(s)
Momento de Replicación del ADN , Replicación del ADN , Animales , Ratones , Cromatina/genética , Replicación del ADN/genética , Heterocromatina/genética , Mamíferos/genética , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Origen de Réplica/genética , Proteínas de Unión a Telómeros/metabolismo
17.
Elife ; 112022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35416772

RESUMEN

RIF1 is a multifunctional protein that plays key roles in the regulation of DNA processing. During repair of DNA double-strand breaks (DSBs), RIF1 functions in the 53BP1-Shieldin pathway that inhibits resection of DNA ends to modulate the cellular decision on which repair pathway to engage. Under conditions of replication stress, RIF1 protects nascent DNA at stalled replication forks from degradation by the DNA2 nuclease. How these RIF1 activities are regulated at the post-translational level has not yet been elucidated. Here, we identified a cluster of conserved ATM/ATR consensus SQ motifs within the intrinsically disordered region (IDR) of mouse RIF1 that are phosphorylated in proliferating B lymphocytes. We found that phosphorylation of the conserved IDR SQ cluster is dispensable for the inhibition of DSB resection by RIF1, but is essential to counteract DNA2-dependent degradation of nascent DNA at stalled replication forks. Therefore, our study identifies a key molecular feature that enables the genome-protective function of RIF1 during DNA replication stress.


Asunto(s)
Roturas del ADN de Doble Cadena , Replicación del ADN , Animales , ADN/metabolismo , Reparación del ADN , Ratones , Fosforilación , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo
18.
Nat Commun ; 12(1): 1929, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33771987

RESUMEN

Leigh syndrome (LS) is a severe manifestation of mitochondrial disease in children and is currently incurable. The lack of effective models hampers our understanding of the mechanisms underlying the neuronal pathology of LS. Using patient-derived induced pluripotent stem cells and CRISPR/Cas9 engineering, we developed a human model of LS caused by mutations in the complex IV assembly gene SURF1. Single-cell RNA-sequencing and multi-omics analysis revealed compromised neuronal morphogenesis in mutant neural cultures and brain organoids. The defects emerged at the level of neural progenitor cells (NPCs), which retained a glycolytic proliferative state that failed to instruct neuronal morphogenesis. LS NPCs carrying mutations in the complex I gene NDUFS4 recapitulated morphogenesis defects. SURF1 gene augmentation and PGC1A induction via bezafibrate treatment supported the metabolic programming of LS NPCs, leading to restored neuronal morphogenesis. Our findings provide mechanistic insights and suggest potential interventional strategies for a rare mitochondrial disease.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Enfermedad de Leigh/genética , Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , Mutación , Neuronas/metabolismo , Organoides/metabolismo , Células Cultivadas , Preescolar , Humanos , Células Madre Pluripotentes Inducidas/citología , Enfermedad de Leigh/metabolismo , Masculino , Metabolómica/métodos , Mitocondrias/genética , Mitocondrias/metabolismo , Morfogénesis/genética , Neuronas/citología , Proteómica/métodos , Análisis de la Célula Individual/métodos , Secuenciación del Exoma
19.
J Cell Biol ; 171(5): 765-71, 2005 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-16330708

RESUMEN

Mre11-Rad50-Nbs1 (MRN) complex involvement in nonhomologous end joining (NHEJ) is controversial. The MRN complex is required for NHEJ in Saccharomyces cerevisiae but not in Schizosaccharomyces pombe. In vertebrates, Mre11, Rad50, and Nbs1 are essential genes, and studies have been limited to cells carrying hypomorphic mutations in Mre11 or Nbs1, which still perform several MRN complex-associated activities. In this study, we analyze the effects of Mre11 loss on the mechanism of vertebrate NHEJ by using a chromatinized plasmid double-strand break (DSB) repair assay in cell-free extracts from Xenopus laevis. Mre11-depleted extracts are able to support efficient NHEJ repair of DSBs regardless of the end structure. Mre11 depletion does not alter the kinetics of end joining or the type and frequency of junctions found in repaired products. Finally, Ku70-independent end-joining events are not affected by Mre11 loss. Our data demonstrate that the MRN complex is not required for efficient and accurate NHEJ-mediated repair of DSBs in this vertebrate system.


Asunto(s)
Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , ADN , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Animales , Antígenos Nucleares/fisiología , Proteínas de Unión al ADN/fisiología , Femenino , Técnicas In Vitro , Autoantígeno Ku , Oocitos/metabolismo , Unión Proteica , Xenopus laevis/genética
20.
J Exp Med ; 217(10)2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32609329

RESUMEN

The establishment of protective humoral immunity is dependent on the ability of mature B cells to undergo antibody gene diversification while adjusting to the physiological stressors induced by activation with the antigen. Mature B cells diversify their antibody genes by class switch recombination (CSR) and somatic hypermutation (SHM), which are both dependent on efficient induction of activation-induced cytidine deaminase (AID). Here, we identified PDGFA-associated protein 1 (Pdap1) as an essential regulator of cellular homeostasis in mature B cells. Pdap1 deficiency leads to sustained expression of the integrated stress response (ISR) effector activating transcription factor 4 (Atf4) and induction of the ISR transcriptional program, increased cell death, and defective AID expression. As a consequence, loss of Pdap1 reduces germinal center B cell formation and impairs CSR and SHM. Thus, Pdap1 protects mature B cells against chronic ISR activation and ensures efficient antibody diversification by promoting their survival and optimal function.


Asunto(s)
Diversidad de Anticuerpos , Linfocitos B/metabolismo , Genes de Inmunoglobulinas/genética , Animales , Linfocitos B/inmunología , Proteína 9 Asociada a CRISPR , Sistemas CRISPR-Cas , Muerte Celular , Diferenciación Celular , Línea Celular , Femenino , Técnica del Anticuerpo Fluorescente , Edición Génica , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA