Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 223
Filtrar
1.
Nature ; 600(7888): 329-333, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34819671

RESUMEN

Efficient humoral responses rely on DNA damage, mutagenesis and error-prone DNA repair. Diversification of B cell receptors through somatic hypermutation and class-switch recombination are initiated by cytidine deamination in DNA mediated by activation-induced cytidine deaminase (AID)1 and by the subsequent excision of the resulting uracils by uracil DNA glycosylase (UNG) and by mismatch repair proteins1-3. Although uracils arising in DNA are accurately repaired1-4, how these pathways are co-opted to generate mutations and double-strand DNA breaks in the context of somatic hypermutation and class-switch recombination is unknown1-3. Here we performed a genome-wide CRISPR-Cas9 knockout screen for genes involved in class-switch recombination and identified FAM72A, a protein that interacts with the nuclear isoform of UNG (UNG2)5 and is overexpressed in several cancers5. We show that the FAM72A-UNG2 interaction controls the levels of UNG2 and that class-switch recombination is defective in Fam72a-/- B cells due to the upregulation of UNG2. Moreover, we show that somatic hypermutation is reduced in Fam72a-/- B cells and that its pattern is skewed upon upregulation of UNG2. Our results are consistent with a model in which FAM72A interacts with UNG2 to control its physiological level by triggering its degradation, regulating the level of uracil excision and thus the balance between error-prone and error-free DNA repair. Our findings have potential implications for tumorigenesis, as reduced levels of UNG2 mediated by overexpression of Fam72a would shift the balance towards mutagenic DNA repair, rendering cells more prone to acquire mutations.


Asunto(s)
Linfocitos B , Reparación de la Incompatibilidad de ADN , Cambio de Clase de Inmunoglobulina , Región de Cambio de la Inmunoglobulina , Mutación , Hipermutación Somática de Inmunoglobulina , Animales , Femenino , Masculino , Ratones , Linfocitos B/metabolismo , Sistemas CRISPR-Cas/genética , Genoma/genética , Cambio de Clase de Inmunoglobulina/genética , Región de Cambio de la Inmunoglobulina/genética , Hipermutación Somática de Inmunoglobulina/genética , Regulación hacia Arriba , Uracilo/metabolismo
2.
Nature ; 600(7888): 324-328, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34819670

RESUMEN

Activation-induced cytidine deaminase (AID) catalyses the deamination of deoxycytidines to deoxyuracils within immunoglobulin genes to induce somatic hypermutation and class-switch recombination1,2. AID-generated deoxyuracils are recognized and processed by subverted base-excision and mismatch repair pathways that ensure a mutagenic outcome in B cells3-6. However, why these DNA repair pathways do not accurately repair AID-induced lesions remains unknown. Here, using a genome-wide CRISPR screen, we show that FAM72A is a major determinant for the error-prone processing of deoxyuracils. Fam72a-deficient CH12F3-2 B cells and primary B cells from Fam72a-/- mice exhibit reduced class-switch recombination and somatic hypermutation frequencies at immunoglobulin and Bcl6 genes, and reduced genome-wide deoxyuracils. The somatic hypermutation spectrum in B cells from Fam72a-/- mice is opposite to that observed in mice deficient in uracil DNA glycosylase 2 (UNG2)7, which suggests that UNG2 is hyperactive in FAM72A-deficient cells. Indeed, FAM72A binds to UNG2, resulting in reduced levels of UNG2 protein in the G1 phase of the cell cycle, coinciding with peak AID activity. FAM72A therefore causes U·G mispairs to persist into S phase, leading to error-prone processing by mismatch repair. By disabling the DNA repair pathways that normally efficiently remove deoxyuracils from DNA, FAM72A enables AID to exert its full effects on antibody maturation. This work has implications in cancer, as the overexpression of FAM72A that is observed in many cancers8 could promote mutagenesis.


Asunto(s)
Linfocitos B , ADN Glicosilasas , Reparación de la Incompatibilidad de ADN , Cambio de Clase de Inmunoglobulina , Proteínas de la Membrana , Mutación , Proteínas de Neoplasias , Hipermutación Somática de Inmunoglobulina , Animales , Femenino , Humanos , Ratones , Linfocitos B/metabolismo , Sistemas CRISPR-Cas , ADN Glicosilasas/antagonistas & inhibidores , ADN Glicosilasas/metabolismo , Epistasis Genética , Células HEK293 , Cambio de Clase de Inmunoglobulina/genética , Región de Cambio de la Inmunoglobulina/genética , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Proteína 2 Homóloga a MutS/genética , Proteína 2 Homóloga a MutS/metabolismo , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Hipermutación Somática de Inmunoglobulina/genética
4.
Proc Natl Acad Sci U S A ; 117(37): 22953-22961, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32868446

RESUMEN

The DNA-dependent protein kinase (DNA-PK), which is composed of the KU heterodimer and the large catalytic subunit (DNA-PKcs), is a classical nonhomologous end-joining (cNHEJ) factor. Naïve B cells undergo class switch recombination (CSR) to generate antibodies with different isotypes by joining two DNA double-strand breaks at different switching regions via the cNHEJ pathway. DNA-PK and the cNHEJ pathway play important roles in the DNA repair phase of CSR. To initiate cNHEJ, KU binds to DNA ends and recruits and activates DNA-PK. Activated DNA-PK phosphorylates DNA-PKcs at the S2056 and T2609 clusters. Loss of T2609 cluster phosphorylation increases radiation sensitivity but whether T2609 phosphorylation has a role in physiological DNA repair remains elusive. Using the DNA-PKcs5A mouse model carrying alanine substitutions at the T2609 cluster, here we show that loss of T2609 phosphorylation of DNA-PKcs does not affect the CSR efficiency. Yet, the CSR junctions recovered from DNA-PKcs5A/5A B cells reveal increased chromosomal translocations, extensive use of distal switch regions (consistent with end resection), and preferential usage of microhomology-all signs of the alternative end-joining pathway. Thus, these results uncover a role of DNA-PKcs T2609 phosphorylation in promoting cNHEJ repair pathway choice during CSR.


Asunto(s)
Proteína Quinasa Activada por ADN/genética , Proteína Quinasa Activada por ADN/metabolismo , Cambio de Clase de Inmunoglobulina/genética , Animales , Linfocitos B/inmunología , Reparación del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Femenino , Reordenamiento Génico , Humanos , Cambio de Clase de Inmunoglobulina/fisiología , Región de Cambio de la Inmunoglobulina/genética , Inmunoglobulinas/genética , Autoantígeno Ku/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Fosforilación , Recombinación Genética/genética , Translocación Genética
5.
Crit Rev Biochem Mol Biol ; 54(4): 333-351, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31509023

RESUMEN

Immunoglobulin (Ig) class switch recombination (CSR) is the gene rearrangement process by which B lymphocytes change the Ig heavy chain constant region to permit a switch of Ig isotype from IgM to IgG, IgA, or IgE. At the DNA level, CSR occurs via generation and joining of DNA double strand breaks (DSBs) at intronic switch regions located just upstream of each of the heavy chain constant regions. Activation-induced deaminase (AID), a B cell specific enzyme, catalyzes cytosine deaminations (converting cytosines to uracils) as the initial DNA lesions that eventually lead to DSBs and CSR. Progress on AID structure integrates very well with knowledge about Ig class switch region nucleic acid structures that are supported by functional studies. It is an ideal time to review what is known about the mechanism of Ig CSR and its relation to somatic hypermutation. There have been many comprehensive reviews on various aspects of the CSR reaction and regulation of AID expression and activity. This review is focused on the relation between AID and switch region nucleic acid structures, with a particular emphasis on R-loops.


Asunto(s)
Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Cambio de Clase de Inmunoglobulina/genética , Región de Cambio de la Inmunoglobulina/genética , Recombinación Genética , Hipermutación Somática de Inmunoglobulina/genética , Animales , Linfocitos B/metabolismo , ADN/genética , ADN/metabolismo , Roturas del ADN de Doble Cadena , Humanos , Inmunoglobulinas/genética , Estructuras R-Loop/genética , Translocación Genética
6.
PLoS Genet ; 15(6): e1007721, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31199803

RESUMEN

B-cell activation yields abundant cell death in parallel to clonal amplification and remodeling of immunoglobulin (Ig) genes by activation-induced deaminase (AID). AID promotes affinity maturation of Ig variable regions and class switch recombination (CSR) in mature B lymphocytes. In the IgH locus, these processes are under control of the 3' regulatory region (3'RR) super-enhancer, a region demonstrated in the mouse to be both transcribed and itself targeted by AID-mediated recombination. Alternatively to CSR, IgH deletions joining Sµ to "like-switch" DNA repeats that flank the 3' super-enhancer can thus accomplish so-called "locus suicide recombination" (LSR) in mouse B-cells. Using an optimized LSR-seq high throughput method, we now show that AID-mediated LSR is evolutionarily conserved and also actively occurs in humans, providing an activation-induced cell death pathway in multiple conditions of B-cell activation. LSR either focuses on the functional IgH allele or is bi-allelic, and its signature is mainly detected when LSR is ongoing while it vanishes from fully differentiated plasma cells or from "resting" blood memory B-cells. Highly diversified breakpoints are distributed either within the upstream (3'RR1) or downstream (3'RR2) copies of the IgH 3' super-enhancer and all conditions activating CSR in vitro also seem to trigger LSR although TLR ligation appeared the most efficient. Molecular analysis of breakpoints and junctions confirms that LSR is AID-dependent and reveals junctional sequences somehow similar to CSR junctions but with increased usage of microhomologies.


Asunto(s)
Linfocitos B/inmunología , Citidina Desaminasa/genética , Región de Cambio de la Inmunoglobulina/genética , Inmunoglobulinas/inmunología , Alelos , Animales , Diferenciación Celular/genética , Citidina Desaminasa/inmunología , Marcación de Gen , Humanos , Región de Cambio de la Inmunoglobulina/inmunología , Tejido Linfoide/inmunología , Ratones , Tonsila Palatina/inmunología , Tonsila Palatina/metabolismo , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología , Secuencias Reguladoras de Ácidos Nucleicos
7.
Proc Natl Acad Sci U S A ; 115(46): E10898-E10906, 2018 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-30373813

RESUMEN

Chimeric antigen receptor (CAR) T cells with a long-lived memory phenotype are correlated with durable, complete remissions in patients with leukemia. However, not all CAR T cell products form robust memory populations, and those that do can induce chronic B cell aplasia in patients. To address these challenges, we previously developed a switchable CAR (sCAR) T cell system that allows fully tunable, on/off control over engineered cellular activity. To further evaluate the platform, we generated and assessed different murine sCAR constructs to determine the factors that afford efficacy, persistence, and expansion of sCAR T cells in a competent immune system. We find that sCAR T cells undergo significant in vivo expansion, which is correlated with potent antitumor efficacy. Most importantly, we show that the switch dosing regimen not only allows control over B cell populations through iterative depletion and repopulation, but that the "rest" period between dosing cycles is the key for induction of memory and expansion of sCAR T cells. These findings introduce rest as a paradigm in enhancing memory and improving the efficacy and persistence of engineered T cell products.


Asunto(s)
Bioingeniería/métodos , Inmunoterapia Adoptiva/métodos , Animales , Antígenos CD19/inmunología , Linfocitos B/inmunología , Citocinas/metabolismo , Citotoxicidad Inmunológica/inmunología , Femenino , Región de Cambio de la Inmunoglobulina/genética , Región de Cambio de la Inmunoglobulina/inmunología , Activación de Linfocitos/fisiología , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Modelos Biológicos , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología
8.
Mol Cell ; 70(4): 650-662.e8, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29731414

RESUMEN

Class switch recombination (CSR) at the immunoglobulin heavy-chain (IgH) locus is associated with the formation of R-loop structures over switch (S) regions. While these often occur co-transcriptionally between nascent RNA and template DNA, we now show that they also form as part of a post-transcriptional mechanism targeting AID to IgH S-regions. This depends on the RNA helicase DDX1 that is also required for CSR in vivo. DDX1 binds to G-quadruplex (G4) structures present in intronic switch transcripts and converts them into S-region R-loops. This in turn targets the cytidine deaminase enzyme AID to S-regions so promoting CSR. Notably R-loop levels over S-regions are diminished by chemical stabilization of G4 RNA or by the expression of a DDX1 ATPase-deficient mutant that acts as a dominant-negative protein to reduce CSR efficiency. In effect, we provide evidence for how S-region transcripts interconvert between G4 and R-loop structures to promote CSR in the IgH locus.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , ARN Helicasas DEAD-box/fisiología , G-Cuádruplex , Cadenas Pesadas de Inmunoglobulina/genética , Región de Cambio de la Inmunoglobulina/genética , ARN/química , Adenosina Trifosfatasas/genética , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Replicación del ADN , Cambio de Clase de Inmunoglobulina , Cadenas Pesadas de Inmunoglobulina/química , Cadenas Pesadas de Inmunoglobulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN/genética , Recombinación Genética
9.
Nature ; 548(7669): 597-601, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28847005

RESUMEN

In two previously described donors, the extracellular domain of LAIR1, a collagen-binding inhibitory receptor encoded on chromosome 19 (ref. 1), was inserted between the V and DJ segments of an antibody. This insertion generated, through somatic mutations, broadly reactive antibodies against RIFINs, a type of variant antigen expressed on the surface of Plasmodium falciparum-infected erythrocytes. To investigate how frequently such antibodies are produced in response to malaria infection, we screened plasma from two large cohorts of individuals living in malaria-endemic regions. Here we report that 5-10% of malaria-exposed individuals, but none of the European blood donors tested, have high levels of LAIR1-containing antibodies that dominate the response to infected erythrocytes without conferring enhanced protection against febrile malaria. By analysing the antibody-producing B cell clones at the protein, cDNA and gDNA levels, we characterized additional LAIR1 insertions between the V and DJ segments and discovered a second insertion modality whereby the LAIR1 exon encoding the extracellular domain and flanking intronic sequences are inserted into the switch region. By exon shuffling, this mechanism leads to the production of bispecific antibodies in which the LAIR1 domain is precisely positioned at the elbow between the VH and CH1 domains. Additionally, in one donor the genomic DNA encoding the VH and CH1 domains was deleted, leading to the production of a camel-like LAIR1-containing antibody. Sequencing of the switch regions of memory B cells from European blood donors revealed frequent templated inserts originating from transcribed genes that, in rare cases, comprised exons with orientations and frames compatible with expression. These results reveal different modalities of LAIR1 insertion that lead to public and dominant antibodies against infected erythrocytes and suggest that insertion of templated DNA represents an additional mechanism of antibody diversification that can be selected in the immune response against pathogens and exploited for B cell engineering.


Asunto(s)
Anticuerpos Antiprotozoarios/química , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Donantes de Sangre , Malaria/inmunología , Mutagénesis Insercional , Plasmodium falciparum/inmunología , Receptores Inmunológicos/genética , Anticuerpos Antiprotozoarios/genética , Antígenos de Protozoos/metabolismo , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Eritrocitos/metabolismo , Eritrocitos/parasitología , Europa (Continente) , Femenino , Genes de las Cadenas Pesadas de las Inmunoglobulinas/genética , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Región de Cambio de la Inmunoglobulina/genética , Memoria Inmunológica , Intrones/genética , Malaria/epidemiología , Malaria/parasitología , Masculino , Plasmodium falciparum/metabolismo , Dominios Proteicos , Receptores Inmunológicos/química , Receptores Inmunológicos/inmunología , Moldes Genéticos , Exones VDJ/genética
10.
J Immunol ; 198(10): 4148-4155, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28416601

RESUMEN

B cells ensure humoral immune responses due to the production of Ag-specific memory B cells and Ab-secreting plasma cells. In secondary lymphoid organs, Ag-driven B cell activation induces terminal maturation and Ig isotype class switch (class switch recombination [CSR]). CSR creates a virtually unique IgH locus in every B cell clone by intrachromosomal recombination between two switch (S) regions upstream of each C region gene. Amount and structural features of CSR junctions reveal valuable information about the CSR mechanism, and analysis of CSR junctions is useful in basic and clinical research studies of B cell functions. To provide an automated tool able to analyze large data sets of CSR junction sequences produced by high-throughput sequencing (HTS), we designed CSReport, a software program dedicated to support analysis of CSR recombination junctions sequenced with a HTS-based protocol (Ion Torrent technology). CSReport was assessed using simulated data sets of CSR junctions and then used for analysis of Sµ-Sα and Sµ-Sγ1 junctions from CH12F3 cells and primary murine B cells, respectively. CSReport identifies junction segment breakpoints on reference sequences and junction structure (blunt-ended junctions or junctions with insertions or microhomology). Besides the ability to analyze unprecedentedly large libraries of junction sequences, CSReport will provide a unified framework for CSR junction studies. Our results show that CSReport is an accurate tool for analysis of sequences from our HTS-based protocol for CSR junctions, thereby facilitating and accelerating their study.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Cambio de Clase de Inmunoglobulina/genética , Recombinación Genética , Programas Informáticos , Linfocitos B/inmunología , Roturas del ADN de Doble Cadena , Isotipos de Inmunoglobulinas/genética , Región de Cambio de la Inmunoglobulina/genética
11.
Nat Commun ; 8: 14244, 2017 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-28176781

RESUMEN

Antibody class-switch DNA recombination (CSR) is initiated by AID-introduced DSBs in the switch (S) regions targeted for recombination, as effected by Ku70/Ku86-mediated NHEJ. Ku-deficient B cells, however, undergo (reduced) CSR through an alternative(A)-NHEJ pathway, which introduces microhomologies in S-S junctions. As microhomology-mediated end-joining requires annealing of single-strand DNA ends, we addressed the contribution of single-strand annealing factors HR Rad52 and translesion DNA polymerase θ to CSR. Compared with their Rad52+/+ counterparts, which display normal CSR, Rad52-/- B cells show increased CSR, fewer intra-Sµ region recombinations, no/minimal microhomologies in S-S junctions, decreased c-Myc/IgH translocations and increased Ku70/Ku86 recruitment to S-region DSB ends. Rad52 competes with Ku70/Ku86 for binding to S-region DSB ends. It also facilitates a Ku-independent DSB repair, which favours intra-S region recombination and mediates, particularly in Ku absence, inter-S-S recombination, as emphasized by the significantly greater CSR reduction in Rad52-/- versus Rad52+/+ B cells on Ku86 knockdown.


Asunto(s)
Reparación del ADN por Unión de Extremidades/inmunología , Cambio de Clase de Inmunoglobulina/genética , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , Reparación del ADN por Recombinación/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Citidina Desaminasa/genética , Citidina Desaminasa/inmunología , Citidina Desaminasa/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades/genética , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/inmunología , ADN Polimerasa Dirigida por ADN/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Cambio de Clase de Inmunoglobulina/inmunología , Región de Cambio de la Inmunoglobulina/genética , Autoantígeno Ku/genética , Autoantígeno Ku/inmunología , Autoantígeno Ku/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño/metabolismo , Proteína Recombinante y Reparadora de ADN Rad52/genética , Proteína Recombinante y Reparadora de ADN Rad52/inmunología , Sulfonamidas , ADN Polimerasa theta
12.
J Immunol ; 197(7): 2930-5, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27559052

RESUMEN

The ∼28-kb 3' regulatory region (3'RR), which is located at the most distal 3' region of the Ig H chain locus, has multiple regulatory functions that control IgH expression, class-switch recombination (CSR), and somatic hypermutation. In this article, we report that deletion of the entire 3'RR in a mouse B cell line that is capable of robust cytokine-dependent CSR to IgA results in reduced, but not abolished, CSR. These data suggest that 3'RR is not absolutely required for CSR and, thus, is not essential for targeting activation-induced cytidine deaminase to S regions, as was suggested. Moreover, replacing 3'RR with a DNA fragment including only its four DNase I hypersensitive sites (lacking the large spacer regions) restores CSR to a level equivalent to or even higher than in wild-type cells, suggesting that the four hypersensitive sites contain most of the CSR-promoting functions of 3'RR. Stimulated cells express abundant germline transcripts, with the presence or absence of 3'RR, providing evidence that 3'RR has a role in promoting CSR that is unique from enhancing S region transcription.


Asunto(s)
Inmunoglobulina G/genética , Cadenas Pesadas de Inmunoglobulina/genética , Región de Cambio de la Inmunoglobulina/genética , Animales , Células Cultivadas , Inmunoglobulina G/inmunología , Cadenas Pesadas de Inmunoglobulina/inmunología , Región de Cambio de la Inmunoglobulina/inmunología , Ratones
13.
Eur J Immunol ; 46(3): 523-30, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26799454

RESUMEN

The mechanism by which the DNA deaminase activation-induced cytidine deaminase (AID) is specifically recruited to repetitive switch region DNA during class switch recombination is still poorly understood. Work over the past decade has revealed a strong link between transcription and RNA polymerase-associated factors in AID recruitment, yet none of these processes satisfactorily explain how AID specificity is affected. Here, we review a recent finding wherein AID is guided to switch regions not by a protein factor but by an RNA moiety, and especially one associated with a noncoding RNA that has been long thought of as being inert. This work explains the long-standing requirement of splicing of noncoding transcripts during class switching, and has implications in both B cell-mediated immunity as well as the underlying pathological syndromes associated with the recombination reaction.


Asunto(s)
Citidina Desaminasa/metabolismo , ADN/metabolismo , Cambio de Clase de Inmunoglobulina/genética , ARN/fisiología , Animales , Citidina Desaminasa/genética , Humanos , Región de Cambio de la Inmunoglobulina/genética , Inmunoglobulinas/genética , Ratones , ARN/genética , Recombinación Genética
14.
Nucleic Acids Res ; 44(2): 718-29, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26527723

RESUMEN

Highly transcribed guanine-run containing sequences, in Saccharomyces cerevisiae, become unstable when topoisomerase I (Top1) is disrupted. Topological changes, such as the formation of extended RNA:DNA hybrids or R-loops or non-canonical DNA structures including G-quadruplexes has been proposed as the major underlying cause of the transcription-linked genome instability. Here, we report that R-loop accumulation at a guanine-rich sequence, which is capable of assembling into the four-stranded G4 DNA structure, is dependent on the level and the orientation of transcription. In the absence of Top1 or RNase Hs, R-loops accumulated to substantially higher extent when guanine-runs were located on the non-transcribed strand. This coincides with the orientation where higher genome instability was observed. However, we further report that there are significant differences between the disruption of RNase Hs and Top1 in regards to the orientation-specific elevation in genome instability at the guanine-rich sequence. Additionally, genome instability in Top1-deficient yeasts is not completely suppressed by removal of negative supercoils and further aggravated by expression of mutant Top1. Together, our data provide a strong support for a function of Top1 in suppressing genome instability at the guanine-run containing sequence that goes beyond preventing the transcription-associated RNA:DNA hybrid formation.


Asunto(s)
ADN-Topoisomerasas de Tipo I/metabolismo , ADN de Hongos/metabolismo , Inestabilidad Genómica , ARN de Hongos/metabolismo , Camptotecina , Replicación del ADN , ADN-Topoisomerasas de Tipo I/genética , ADN de Hongos/química , ADN de Hongos/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , G-Cuádruplex , Prueba de Complementación Genética , Guanina/química , Guanina/metabolismo , Región de Cambio de la Inmunoglobulina/genética , Hibridación de Ácido Nucleico , ARN de Hongos/genética , Recombinación Genética , Ribonucleasa H/genética , Ribonucleasa H/metabolismo , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Transcripción Genética
15.
PLoS One ; 10(8): e0134397, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26267846

RESUMEN

Activation-induced cytidine deaminase (AID) is essential for class switch recombination (CSR) and somatic hypermutation (SHM) of Ig genes. The C terminus of AID is required for CSR but not for SHM, but the reason for this is not entirely clear. By retroviral transduction of mutant AID proteins into aid-/- mouse splenic B cells, we show that 4 amino acids within the C terminus of mouse AID, when individually mutated to specific amino acids (R190K, A192K, L196S, F198S), reduce CSR about as much or more than deletion of the entire C terminal 10 amino acids. Similar to ΔAID, the substitutions reduce binding of UNG to Ig Sµ regions and some reduce binding of Msh2, both of which are important for introducing S region DNA breaks. Junctions between the IgH donor switch (S)µ and acceptor Sα regions from cells expressing ΔAID or the L196S mutant show increased microhomology compared to junctions in cells expressing wild-type AID, consistent with problems during CSR and the use of alternative end-joining, rather than non-homologous end-joining (NHEJ). Unlike deletion of the AID C terminus, 3 of the substitution mutants reduce DNA double-strand breaks (DSBs) detected within the Sµ region in splenic B cells undergoing CSR. Cells expressing these 3 substitution mutants also have greatly reduced mutations within unrearranged Sµ regions, and they decrease with time after activation. These results might be explained by increased error-free repair, but as the C terminus has been shown to be important for recruitment of NHEJ proteins, this appears unlikely. We hypothesize that Sµ DNA breaks in cells expressing these C terminus substitution mutants are poorly repaired, resulting in destruction of Sµ segments that are deaminated by these mutants. This could explain why these mutants cannot undergo CSR.


Asunto(s)
Citidina Desaminasa/genética , Cambio de Clase de Inmunoglobulina/genética , Región de Cambio de la Inmunoglobulina/genética , Inmunoglobulinas/genética , Recombinación Genética , Sustitución de Aminoácidos/genética , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Citidina Desaminasa/inmunología , ADN/genética , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades/genética , Humanos , Ratones , Ratones Noqueados , Mutación Missense
16.
Mol Immunol ; 67(2 Pt B): 524-31, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26277278

RESUMEN

Vertebrates developed immunoglobulin heavy chain (IgH) class switch recombination (CSR) to express different IgH constant regions. Most double-strand breaks for Ig CSR occur within the repetitive portion of the switch regions located upstream of each set of constant domain exons for the Igγ, Igα or Igϵ heavy chain. Unlike mammalian switch regions, Xenopus switch regions do not have a high G-density on the non-template DNA strand. In previous studies, when Xenopus Sµ DNA was moved to the genome of mice, it is able to support substantial CSR when it is used to replace the murine Sγ1 region. Here, we tested both the 2kb repetitive portion and the 4.6 kb full-length portions of the Xenopus Sµ in both their natural (forward) orientation relative to the constant domain exons, as well as the opposite (reverse) orientation. Consistent with previous work, we find that the 4.6 kb full-length Sµ mediates similar levels of CSR in both the forward and reverse orientations. Whereas, the forward orientation of the 2kb portion can restore the majority of the CSR level of the 4.6 kb full-length Sµ, the reverse orientation poorly supports R-looping and no CSR. The forward orientation of the 2kb repetitive portion has more GG dinucleotides on the non-template strand than the reverse orientation. The correlation of R-loop formation with CSR efficiency, as demonstrated in the 2kb repetitive fragment of the Xenopus switch region, confirms a role played by R-looping in CSR that appears to be conserved through evolution.


Asunto(s)
Cambio de Clase de Inmunoglobulina/inmunología , Cadenas Pesadas de Inmunoglobulina/inmunología , Región de Cambio de la Inmunoglobulina/inmunología , Cadenas mu de Inmunoglobulina/inmunología , Secuencias Repetitivas de Aminoácido , Xenopus/inmunología , Secuencias de Aminoácidos , Animales , Cadenas Pesadas de Inmunoglobulina/química , Región de Cambio de la Inmunoglobulina/genética , Cadenas mu de Inmunoglobulina/química , Transcripción Genética
17.
PLoS Genet ; 11(5): e1005240, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-26000965

RESUMEN

To generate highly specific and adapted immune responses, B cells diversify their antibody repertoire through mechanisms involving the generation of programmed DNA damage. Somatic hypermutation (SHM) and class switch recombination (CSR) are initiated by the recruitment of activation-induced cytidine deaminase (AID) to immunoglobulin loci and by the subsequent generation of DNA lesions, which are differentially processed to mutations during SHM or to double-stranded DNA break intermediates during CSR. The latter activate the DNA damage response and mobilize multiple DNA repair factors, including Parp1 and Parp2, to promote DNA repair and long-range recombination. We examined the contribution of Parp3 in CSR and SHM. We find that deficiency in Parp3 results in enhanced CSR, while SHM remains unaffected. Mechanistically, this is due to increased occupancy of AID at the donor (Sµ) switch region. We also find evidence of increased levels of DNA damage at switch region junctions and a bias towards alternative end joining in the absence of Parp3. We propose that Parp3 plays a CSR-specific role by controlling AID levels at switch regions during CSR.


Asunto(s)
Regulación de la Expresión Génica , Cambio de Clase de Inmunoglobulina/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Animales , Linfocitos B/metabolismo , Secuencia de Bases , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN , Sitios Genéticos , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Región de Cambio de la Inmunoglobulina/genética , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Poli(ADP-Ribosa) Polimerasas/genética , Recombinación Genética , Hipermutación Somática de Inmunoglobulina/genética
18.
PLoS Genet ; 10(12): e1004839, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25473964

RESUMEN

G-quadruplex or G4 DNA is a non-B secondary DNA structure that comprises a stacked array of guanine-quartets. Cellular processes such as transcription and replication can be hindered by unresolved DNA secondary structures potentially endangering genome maintenance. As G4-forming sequences are highly frequent throughout eukaryotic genomes, it is important to define what factors contribute to a G4 motif becoming a hotspot of genome instability. Using a genetic assay in Saccharomyces cerevisiae, we previously demonstrated that a potential G4-forming sequence derived from a guanine-run containing immunoglobulin switch Mu (Sµ) region becomes highly unstable when actively transcribed. Here we describe assays designed to survey spontaneous genome rearrangements initiated at the Sµ sequence in the context of large genomic areas. We demonstrate that, in the absence of Top1, a G4 DNA-forming sequence becomes a strong hotspot of gross chromosomal rearrangements and loss of heterozygosity associated with mitotic recombination within the ∼ 20 kb or ∼ 100 kb regions of yeast chromosome V or III, respectively. Transcription confers a critical strand bias since genome rearrangements at the G4-forming Sµ are elevated only when the guanine-runs are located on the non-transcribed strand. The direction of replication and transcription, when in a head-on orientation, further contribute to the elevated genome instability at a potential G4 DNA-forming sequence. The implications of our identification of Top1 as a critical factor in suppression of instability associated with potential G4 DNA-forming sequences are discussed.


Asunto(s)
ADN-Topoisomerasas de Tipo I/fisiología , G-Cuádruplex , Inestabilidad Genómica , Saccharomyces cerevisiae , Transcripción Genética , Eliminación de Gen , Guanina/metabolismo , Región de Cambio de la Inmunoglobulina/genética , Secuencias Invertidas Repetidas , Organismos Modificados Genéticamente , Recombinación Genética , Saccharomyces cerevisiae/genética , Telómero/genética , Telómero/metabolismo
19.
J Exp Med ; 211(6): 1011-8, 2014 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-24799500

RESUMEN

Fanconi anemia is a rare genetic disorder that can lead to bone marrow failure, congenital abnormalities, and increased risk for leukemia and cancer. Cells with loss-of-function mutations in the FANC pathway are characterized by chromosome fragility, altered mutability, and abnormal regulation of the nonhomologous end-joining (NHEJ) pathway. Somatic hypermutation (SHM) and immunoglobulin (Ig) class switch recombination (CSR) enable B cells to produce high-affinity antibodies of various isotypes. Both processes are initiated after the generation of dG:dU mismatches by activation-induced cytidine deaminase. Whereas SHM involves an error-prone repair process that introduces novel point mutations into the Ig gene, the mismatches generated during CSR are processed to create double-stranded breaks (DSBs) in DNA, which are then repaired by the NHEJ pathway. As several lines of evidence suggest a possible role for the FANC pathway in SHM and CSR, we analyzed both processes in B cells derived from Fanca(-/-) mice. Here we show that Fanca is required for the induction of transition mutations at A/T residues during SHM and that despite globally normal CSR function in splenic B cells, Fanca is required during CSR to stabilize duplexes between pairs of short microhomology regions, thereby impeding short-range recombination downstream of DSB formation.


Asunto(s)
Linfocitos B/metabolismo , Proteína del Grupo de Complementación A de la Anemia de Fanconi/metabolismo , Cambio de Clase de Inmunoglobulina/genética , Mutación Puntual , Hipermutación Somática de Inmunoglobulina/genética , Animales , Linfocitos B/inmunología , Secuencia de Bases , Western Blotting , Línea Celular , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Proteína del Grupo de Complementación A de la Anemia de Fanconi/deficiencia , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación G de la Anemia de Fanconi/deficiencia , Proteína del Grupo de Complementación G de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación G de la Anemia de Fanconi/metabolismo , Humanos , Región de Cambio de la Inmunoglobulina/genética , Ratones de la Cepa 129 , Ratones Noqueados , Datos de Secuencia Molecular , Proteína 2 Homóloga a MutS/metabolismo , Reacción en Cadena de la Polimerasa , Recombinación Genética
20.
Proc Natl Acad Sci U S A ; 111(11): E1016-24, 2014 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-24591630

RESUMEN

Activation-induced cytidine deaminase (AID) is essential to class-switch recombination (CSR) and somatic hypermutation (SHM) in both V region SHM and S region SHM (s-SHM). Uracil DNA glycosylase (UNG), a member of the base excision repair (BER) complex, is required for CSR. Strikingly, however, UNG deficiency causes augmentation of SHM, suggesting involvement of distinct functions of UNG in SHM and CSR. Here, we show that noncanonical scaffold functions of UNG regulate s-SHM negatively and CSR positively. The s-SHM suppressive function of UNG is attributed to the recruitment of faithful BER components at the cleaved DNA locus, with competition against error-prone polymerases. By contrast, the CSR-promoting function of UNG enhances AID-dependent S-S synapse formation by recruiting p53-binding protein 1 and DNA-dependent protein kinase, catalytic subunit. Several loss-of-catalysis mutants of UNG discriminated CSR-promoting activity from s-SHM suppressive activity. Taken together, the noncanonical function of UNG regulates the steps after AID-induced DNA cleavage: error-prone repair suppression in s-SHM and end-joining promotion in CSR.


Asunto(s)
Citidina Desaminasa/metabolismo , Cambio de Clase de Inmunoglobulina/inmunología , Región de Cambio de la Inmunoglobulina/genética , Modelos Moleculares , Hipermutación Somática de Inmunoglobulina/inmunología , Uracil-ADN Glicosidasa/metabolismo , Animales , Inmunoprecipitación de Cromatina , Citidina Desaminasa/genética , Reparación del ADN por Unión de Extremidades/inmunología , Cartilla de ADN/genética , Citometría de Flujo , Fluorescencia , Proteínas Fluorescentes Verdes/genética , Inmunoprecipitación , Ratones , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Hipermutación Somática de Inmunoglobulina/genética , Uracil-ADN Glicosidasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...