Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Nat Immunol ; 12(2): 160-6, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21186367

RESUMEN

During immunoglobulin class-switch recombination (CSR), the cytidine deaminase AID induces double-strand breaks into transcribed, repetitive DNA elements called switch sequences. The mechanism that promotes the binding of AID specifically to switch regions remains to be elucidated. Here we used a proteomic screen with in vivo biotinylation of AID to identify the splicing regulator PTBP2 as a protein that interacts with AID. Knockdown of PTBP2 mediated by short hairpin RNA in B cells led to a decrease in binding of AID to transcribed switch regions, which resulted in considerable impairment of CSR. PTBP2 is thus an effector of CSR that promotes the binding of AID to switch-region DNA.


Asunto(s)
Citidina Desaminasa/metabolismo , ADN/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Citidina Desaminasa/genética , Citidina Desaminasa/inmunología , ADN/genética , Cambio de Clase de Inmunoglobulina/genética , Región de Cambio de la Inmunoglobulina/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Proteína de Unión al Tracto de Polipirimidina/genética , Proteína de Unión al Tracto de Polipirimidina/inmunología , Unión Proteica/genética , ARN Interferente Pequeño/genética , Activación Transcripcional/genética , Activación Transcripcional/inmunología , Transgenes/genética
2.
J Biol Chem ; 296: 100625, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33831416

RESUMEN

Class switch recombination (CSR) is the process by which B cells switch production from IgM/IgD to other immunoglobulin isotypes, enabling them to mount an effective immune response against pathogens. Timely resolution of CSR prevents damage due to an uncontrolled and prolonged immune response. While many positive regulators of CSR have been described, negative regulators of CSR are relatively unknown. Using an shRNA library screen targeting more than 28,000 genes in a mouse B cell line, we have identified a novel, uncharacterized protein of 82kD (KIAA1841, NM_027860), which we have named SANBR (SANT and BTB domain regulator of CSR), as a negative regulator of CSR. The purified, recombinant BTB domain of SANBR exhibited characteristic properties such as homodimerization and interaction with corepressor proteins, including HDAC and SMRT. Overexpression of SANBR inhibited CSR in primary mouse splenic B cells, and inhibition of CSR is dependent on the BTB domain while the SANT domain is largely dispensable. Thus, we have identified a new member of the BTB family that serves as a negative regulator of CSR. Future investigations to identify transcriptional targets of SANBR in B cells will reveal further insights into the specific mechanisms by which SANBR regulates CSR as well as fundamental gene regulatory activities of this protein.


Asunto(s)
Dominio BTB-POZ , Proteínas de Unión al ADN/metabolismo , Cambio de Clase de Inmunoglobulina , Linfoma de Células B/patología , Recombinación Genética , Secuencia de Aminoácidos , Animales , Linfocitos B/metabolismo , Linfocitos B/patología , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Femenino , Humanos , Linfoma de Células B/genética , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño/genética , Homología de Secuencia
3.
J Immunol ; 204(1): 13-22, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31757865

RESUMEN

Activation-induced cytidine deaminase (AID) generates U:G mismatches in Ig genes that can be converted into untemplated mutations during somatic hypermutation or DNA double-strand breaks during class switch recombination (CSR). Null mutations in UNG and MSH2 demonstrate the complementary roles of the base excision repair (BER) and mismatch repair pathways, respectively, in CSR. Phosphorylation of AID at serine 38 was previously hypothesized to regulate BER during CSR, as the AID phosphorylation mutant, AID(S38A), cannot interact with APE1, a BER protein. Consistent with these findings, we observe a complete block in CSR in AIDS38A/S38AMSH2-/- mouse B cells that correlates with an impaired mutation frequency at 5'Sµ. Similarly, somatic hypermutation is almost negligible at the JH4 intron in AIDS38A/S38AMSH2-/- mouse B cells, and, consistent with this, NP-specific affinity maturation in AIDS38A/S38AMSH2-/- mice is not significantly elevated in response to NP-CGG immunization. Surprisingly, AIDS38A/S38AUNG-/- mouse B cells also cannot complete CSR or affinity maturation despite accumulating significant mutations in 5'Sµ as well as the JH4 intron. These data identify a novel role for phosphorylation of AID at serine 38 in mismatch repair-dependent CSR and affinity maturation.


Asunto(s)
Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Reparación de la Incompatibilidad de ADN/genética , Cambio de Clase de Inmunoglobulina/genética , Hipermutación Somática de Inmunoglobulina/genética , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Roturas del ADN de Doble Cadena , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Femenino , Genes de Inmunoglobulinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína 2 Homóloga a MutS/genética , Fosforilación , Recombinación Genética , Uracil-ADN Glicosidasa/genética
4.
J Immunol ; 193(1): 252-7, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24879790

RESUMEN

The DNA deaminase activation-induced cytidine deaminase (AID) initiates somatic hypermutation (SHM) and class switch recombination (CSR) by deaminating cytidines to uridines at V region (V) genes and switch (S) regions. The mechanism by which AID is recruited to V genes and S region DNA is poorly understood. In this study, we used the CH12 B lymphoma line to demonstrate that, although S regions can efficiently recruit AID and undergo mutations and deletions, AID neither binds to nor mutates the V gene, thus clearly demonstrating intraimmunoglobulin locus specificity. Depletion of the RNA-binding protein polypyrimidine tract binding protein-2, previously shown to promote recruitment of AID to S regions, enables stable association of AID with the V gene. Surprisingly, AID binding to the V gene does not induce SHM. These results unmask a striking lack of correlation between AID binding and its mutator activity, providing evidence for the presence of factors required downstream of AID binding to effect SHM. Furthermore, our findings suggest that S regions are preferred targets for AID and, aided by polypyrimidine tract binding protein-2, act as "sinks" to sequester AID activity from other genomic regions.


Asunto(s)
Secuencia de Bases , Citidina Desaminasa/inmunología , ADN/inmunología , Proteínas del Tejido Nervioso/inmunología , Proteína de Unión al Tracto de Polipirimidina/inmunología , Eliminación de Secuencia/inmunología , Animales , Línea Celular Tumoral , Citidina Desaminasa/genética , ADN/genética , Ratones , Proteínas del Tejido Nervioso/genética , Proteína de Unión al Tracto de Polipirimidina/genética , Unión Proteica
5.
Elife ; 112022 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-36190107

RESUMEN

During the development of humoral immunity, activated B lymphocytes undergo vigorous proliferative, transcriptional, metabolic, and DNA remodeling activities; hence, their genomes are constantly exposed to an onslaught of genotoxic agents and processes. Branched DNA intermediates generated during replication and recombinational repair pose genomic threats if left unresolved and so, they must be eliminated by structure-selective endonucleases to preserve the integrity of these DNA transactions for the faithful duplication and propagation of genetic information. To investigate the role of two such enzymes, GEN1 and MUS81, in B cell biology, we established B-cell conditional knockout mouse models and found that deletion of GEN1 and MUS81 in early B-cell precursors abrogates the development and maturation of B-lineage cells while the loss of these enzymes in mature B cells inhibit the generation of robust germinal centers. Upon activation, these double-null mature B lymphocytes fail to proliferate and survive while exhibiting transcriptional signatures of p53 signaling, apoptosis, and type I interferon response. Metaphase spreads of these endonuclease-deficient cells showed severe and diverse chromosomal abnormalities, including a preponderance of chromosome breaks, consistent with a defect in resolving recombination intermediates. These observations underscore the pivotal roles of GEN1 and MUS81 in safeguarding the genome to ensure the proper development and proliferation of B lymphocytes.


Asunto(s)
Endonucleasas , Interferón Tipo I , Animales , Ratones , Linfocitos B/metabolismo , ADN , Endonucleasas/genética , Endonucleasas/metabolismo , Resolvasas de Unión Holliday/genética , Resolvasas de Unión Holliday/metabolismo , Interferón Tipo I/metabolismo , Proteína p53 Supresora de Tumor , Genoma
6.
Adv Immunol ; 122: 1-57, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24507154

RESUMEN

Upon encountering antigens, mature IgM-positive B lymphocytes undergo class-switch recombination (CSR) wherein exons encoding the default Cµ constant coding gene segment of the immunoglobulin (Ig) heavy-chain (Igh) locus are excised and replaced with a new constant gene segment (referred to as "Ch genes", e.g., Cγ, Cɛ, or Cα). The B cell thereby changes from expressing IgM to one producing IgG, IgE, or IgA, with each antibody isotype having a different effector function during an immune reaction. CSR is a DNA deletional-recombination reaction that proceeds through the generation of DNA double-strand breaks (DSBs) in repetitive switch (S) sequences preceding each Ch gene and is completed by end-joining between donor Sµ and acceptor S regions. CSR is a multistep reaction requiring transcription through S regions, the DNA cytidine deaminase AID, and the participation of several general DNA repair pathways including base excision repair, mismatch repair, and classical nonhomologous end-joining. In this review, we discuss our current understanding of how transcription through S regions generates substrates for AID-mediated deamination and how AID participates not only in the initiation of CSR but also in the conversion of deaminated residues into DSBs. Additionally, we review the multiple processes that regulate AID expression and facilitate its recruitment specifically to the Ig loci, and how deregulation of AID specificity leads to oncogenic translocations. Finally, we summarize recent data on the potential role of AID in the maintenance of the pluripotent stem cell state during epigenetic reprogramming.


Asunto(s)
Citidina Desaminasa/metabolismo , Daño del ADN/inmunología , Reparación del ADN/inmunología , Cambio de Clase de Inmunoglobulina/genética , Transcripción Genética/inmunología , Recombinación V(D)J/inmunología , Animales , Linfocitos B/enzimología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Daño del ADN/genética , Reparación del ADN/genética , Humanos , Cambio de Clase de Inmunoglobulina/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Transgénicos , Recombinación Genética/genética , Recombinación Genética/inmunología , Hipermutación Somática de Inmunoglobulina/genética , Hipermutación Somática de Inmunoglobulina/inmunología , Linfocitos T/enzimología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Recombinación V(D)J/genética
7.
Nat Struct Mol Biol ; 18(1): 75-9, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21131982

RESUMEN

Immunoglobulin heavy chain (Igh locus) class-switch recombination (CSR) requires targeted introduction of DNA double strand breaks (DSBs) into repetitive 'switch'-region DNA elements in the Igh locus and subsequent ligation between distal DSBs. Both canonical nonhomologous end joining (C-NHEJ) that seals DNA ends with little or no homology and a poorly defined alternative end joining (A-NHEJ, also known as alt-NHEJ) process that requires microhomology ends for ligation have been implicated in CSR. Here, we show that the DNA end-processing factor CtIP is required for microhomology-directed A-NHEJ during CSR. Additionally, we demonstrate that microhomology joins that are enriched upon depletion of the C-NHEJ component Ku70 require CtIP. Finally, we show that CtIP binds to switch-region DNA in a fashion dependent on activation-induced cytidine deaminase. Our results establish CtIP as a bona fide component of microhomology-dependent A-NHEJ and unmask a hitherto unrecognized physiological role of microhomology-mediated end joining in a C-NHEJ-proficient environment.


Asunto(s)
Proteínas Portadoras/fisiología , Proteínas de Ciclo Celular/fisiología , Cambio de Clase de Inmunoglobulina/fisiología , Recombinación Genética , Animales , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Línea Celular , ADN/química , Roturas del ADN de Doble Cadena , Técnicas de Silenciamiento del Gen , Cambio de Clase de Inmunoglobulina/genética , Ratones , Modelos Genéticos , ARN Interferente Pequeño/fisiología , Homología de Secuencia de Ácido Nucleico
8.
J Immunol ; 181(1): 276-87, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18566393

RESUMEN

Class switch DNA recombination (CSR) from IgM to IgG and IgA is crucial for antiviral immunity. Follicular B cells undergo CSR upon engagement of CD40 by CD40 ligand on CD4+ T cells. This T cell-dependent pathway requires 5-7 days, which is too much of a delay to block quickly replicating pathogens. To compensate for this limitation, extrafollicular B cells rapidly undergo CSR through a T cell-independent pathway that involves innate Ag receptors of the TLR family. We found that a subset of upper respiratory mucosa B cells expressed TLR3 and responded to viral dsRNA, a cognate TLR3 ligand. In the presence of dsRNA, mucosal B cells activated NF-kappaB, a transcription factor critical for CSR. Activation of NF-kappaB required TRIF (Toll/IL-1R domain-containing protein inducing IFN-beta), a canonical TLR3 adapter protein, and caused germline transcription of downstream CH genes as well as expression of AID (activation-induced cytidine deaminase), a DNA-editing enzyme essential for CSR. Subsequent IgG and IgA production was enhanced by BAFF (B cell-activating factor of the TNF family), an innate mediator released by TLR3-expressing mucosal dendritic cells. Indeed, these innate immune cells triggered IgG and IgA responses upon exposure to dsRNA. By showing active TLR3 signaling and ongoing CSR in upper respiratory mucosa B cells from patients with CD40 signaling defects, our findings indicate that viral dsRNA may initiate frontline IgG and IgA responses through an innate TLR3-dependent pathway involving BAFF.


Asunto(s)
Factor Activador de Células B/metabolismo , Linfocitos B/inmunología , Cambio de Clase de Inmunoglobulina/inmunología , Inmunoglobulinas/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 3/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Linfocitos B/citología , Linfocitos B/metabolismo , Antígenos CD40/inmunología , Proliferación Celular , Células Cultivadas , Citidina Desaminasa/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Cambio de Clase de Inmunoglobulina/genética , Inmunoglobulinas/genética , Inmunoglobulinas/metabolismo , Membrana Mucosa/citología , Membrana Mucosa/inmunología , ARN Bicatenario/genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA