Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(10): 2193-2207.e19, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37098343

RESUMEN

Somatic hypermutation (SHM), initiated by activation-induced cytidine deaminase (AID), generates mutations in the antibody-coding sequence to allow affinity maturation. Why these mutations intrinsically focus on the three nonconsecutive complementarity-determining regions (CDRs) remains enigmatic. Here, we found that predisposition mutagenesis depends on the single-strand (ss) DNA substrate flexibility determined by the mesoscale sequence surrounding AID deaminase motifs. Mesoscale DNA sequences containing flexible pyrimidine-pyrimidine bases bind effectively to the positively charged surface patches of AID, resulting in preferential deamination activities. The CDR hypermutability is mimicable in in vitro deaminase assays and is evolutionarily conserved among species using SHM as a major diversification strategy. We demonstrated that mesoscale sequence alterations tune the in vivo mutability and promote mutations in an otherwise cold region in mice. Our results show a non-coding role of antibody-coding sequence in directing hypermutation, paving the way for the synthetic design of humanized animal models for optimal antibody discovery and explaining the AID mutagenesis pattern in lymphoma.


Asunto(s)
Citidina Desaminasa , Hipermutación Somática de Inmunoglobulina , Animales , Ratones , Anticuerpos/genética , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , ADN/genética , ADN de Cadena Simple , Mutación , Evolución Molecular , Regiones Determinantes de Complementariedad/genética , Motivos de Nucleótidos
2.
Mol Cell ; 84(7): 1206-1223.e15, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38423014

RESUMEN

Appropriate DNA end synapsis, regulated by core components of the synaptic complex including KU70-KU80, LIG4, XRCC4, and XLF, is central to non-homologous end joining (NHEJ) repair of chromatinized DNA double-strand breaks (DSBs). However, it remains enigmatic whether chromatin modifications can influence the formation of NHEJ synaptic complex at DNA ends, and if so, how this is achieved. Here, we report that the mitotic deacetylase complex (MiDAC) serves as a key regulator of DNA end synapsis during NHEJ repair in mammalian cells. Mechanistically, MiDAC removes combinatorial acetyl marks on histone H2A (H2AK5acK9ac) around DSB-proximal chromatin, suppressing hyperaccumulation of bromodomain-containing protein BRD4 that would otherwise undergo liquid-liquid phase separation with KU80 and prevent the proper installation of LIG4-XRCC4-XLF onto DSB ends. This study provides mechanistic insight into the control of NHEJ synaptic complex assembly by a specific chromatin signature and highlights the critical role of H2A hypoacetylation in restraining unscheduled compartmentalization of DNA repair machinery.


Asunto(s)
Cromatina , Proteínas Nucleares , Animales , Cromatina/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , ADN/genética , Reparación del ADN por Unión de Extremidades , Histonas/genética , Histonas/metabolismo , Emparejamiento Cromosómico , Autoantígeno Ku/genética , Autoantígeno Ku/metabolismo , Mamíferos/metabolismo
3.
Cell ; 163(4): 947-59, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26593423

RESUMEN

RAG initiates antibody V(D)J recombination in developing lymphocytes by generating "on-target" DNA breaks at matched pairs of bona fide recombination signal sequences (RSSs). We employ bait RAG-generated breaks in endogenous or ectopically inserted RSS pairs to identify huge numbers of RAG "off-target" breaks. Such breaks occur at the simple CAC motif that defines the RSS cleavage site and are largely confined within convergent CTCF-binding element (CBE)-flanked loop domains containing bait RSS pairs. Marked orientation dependence of RAG off-target activity within loops spanning up to 2 megabases implies involvement of linear tracking. In this regard, major RAG off-targets in chromosomal translocations occur as convergent RSS pairs at enhancers within a loop. Finally, deletion of a CBE-based IgH locus element disrupts V(D)J recombination domains and, correspondingly, alters RAG on- and off-target distributions within IgH. Our findings reveal how RAG activity is developmentally focused and implicate mechanisms by which chromatin domains harness biological processes within them.


Asunto(s)
Cromosomas de los Mamíferos/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Recombinación V(D)J , Animales , Factor de Unión a CCCTC , Cromosomas de los Mamíferos/química , Proteínas de Unión al ADN/metabolismo , Genes myc , Genoma , Secuenciación de Nucleótidos de Alto Rendimiento , Proteínas de Homeodominio/metabolismo , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Linfoma/genética , Ratones , Motivos de Nucleótidos , Proteínas Represoras/metabolismo , Análisis de Secuencia de ADN , Translocación Genética
4.
Cell ; 163(5): 1124-1137, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26582132

RESUMEN

In activated B lymphocytes, AID initiates antibody variable (V) exon somatic hypermutation (SHM) for affinity maturation in germinal centers (GCs) and IgH switch (S) region DNA breaks (DSBs) for class-switch recombination (CSR). To resolve long-standing questions, we have developed an in vivo assay to study AID targeting of passenger sequences replacing a V exon. First, we find AID targets SHM hotspots within V exon and S region passengers at similar frequencies and that the normal SHM process frequently generates deletions, indicating that SHM and CSR employ the same mechanism. Second, AID mutates targets in diverse non-Ig passengers in GC B cells at levels similar to those of V exons, definitively establishing the V exon location as "privileged" for SHM. Finally, Peyer's patch GC B cells generate a reservoir of V exons that are highly mutated before selection for affinity maturation. We discuss the implications of these findings for harnessing antibody diversification mechanisms.


Asunto(s)
Linfocitos B/metabolismo , Citidina Desaminasa/genética , Cambio de Clase de Inmunoglobulina , Hipermutación Somática de Inmunoglobulina , Recombinación V(D)J , Animales , Humanos , Ratones , Mutación , Globinas beta/genética
5.
Cell ; 159(7): 1538-48, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25483776

RESUMEN

Activation-induced cytidine deaminase (AID) initiates both somatic hypermutation (SHM) for antibody affinity maturation and DNA breakage for antibody class switch recombination (CSR) via transcription-dependent cytidine deamination of single-stranded DNA targets. Though largely specific for immunoglobulin genes, AID also acts on a limited set of off-targets, generating oncogenic translocations and mutations that contribute to B cell lymphoma. How AID is recruited to off-targets has been a long-standing mystery. Based on deep GRO-seq studies of mouse and human B lineage cells activated for CSR or SHM, we report that most robust AID off-target translocations occur within highly focal regions of target genes in which sense and antisense transcription converge. Moreover, we found that such AID-targeting "convergent" transcription arises from antisense transcription that emanates from super-enhancers within sense transcribed gene bodies. Our findings provide an explanation for AID off-targeting to a small subset of mostly lineage-specific genes in activated B cells.


Asunto(s)
Citidina Desaminasa/metabolismo , Elementos de Facilitación Genéticos , Inestabilidad Genómica , Transcripción Genética , Animales , Linfocitos B/metabolismo , Humanos , Cambio de Clase de Inmunoglobulina , Ratones , Sitio de Iniciación de la Transcripción
6.
Trends Biochem Sci ; 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38614818

RESUMEN

Activation-induced cytidine deaminase (AID) initiates somatic hypermutation (SHM) by introducing base substitutions into antibody genes, a process enabling antibody affinity maturation in immune response. How a mutator is tamed to precisely and safely generate programmed DNA lesions in a physiological process remains unsettled, as its dysregulation drives lymphomagenesis. Recent research has revealed several hidden features of AID-initiated mutagenesis: preferential activity on flexible DNA substrates, restrained activity within chromatin loop domains, unique DNA repair factors to differentially decode AID-caused lesions, and diverse consequences of aberrant deamination. Here, we depict the multifaceted regulation of AID activity with a focus on emerging concepts/factors and discuss their implications for the design of base editors (BEs) that install somatic mutations to correct deleterious genomic variants.

7.
Cell ; 152(3): 417-29, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23374339

RESUMEN

Chromosomal translocations involving antigen receptor loci are common in lymphoid malignancies. Translocations require DNA double-strand breaks (DSBs) at two chromosomal sites, their physical juxtaposition, and their fusion by end-joining. Ability of lymphocytes to generate diverse repertoires of antigen receptors and effector antibodies derives from programmed genomic alterations that produce DSBs. We discuss these lymphocyte-specific processes, with a focus on mechanisms that provide requisite DSB target specificity and mechanisms that suppress DSB translocation. We also discuss recent work that provides new insights into DSB repair pathways and the influences of three-dimensional genome organization on physiological processes and cancer genomes.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Inestabilidad Genómica , Linfocitos/metabolismo , Recombinación V(D)J , Animales , Humanos , Linfocitos/inmunología , Linfoma/genética , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos T/genética , Translocación Genética
8.
Trends Immunol ; 45(3): 167-176, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38402044

RESUMEN

Antibody-coding genes accumulate somatic mutations to achieve antibody affinity maturation. Genetic dissection using various mouse models has shown that intrinsic hypermutations occur preferentially and are predisposed in the DNA region encoding antigen-contacting residues. The molecular basis of nonrandom/preferential mutations is a long-sought question in the field. Here, we summarize recent findings on how single-strand (ss)DNA flexibility facilitates activation-induced cytidine deaminase (AID) activity and fine-tunes the mutation rates at a mesoscale within the antibody variable domain exon. We propose that antibody coding sequences are selected based on mutability during the evolution of adaptive immunity and that DNA mechanics play a noncoding role in the genome. The mechanics code may also determine other cellular DNA metabolism processes, which awaits future investigation.


Asunto(s)
Genes de Inmunoglobulinas , Hipermutación Somática de Inmunoglobulina , Animales , Ratones , Hipermutación Somática de Inmunoglobulina/genética , Mutación , ADN , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo
9.
EMBO J ; 41(11): e109324, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35471583

RESUMEN

In activated B cells, activation-induced cytidine deaminase (AID) generates programmed DNA lesions required for antibody class switch recombination (CSR), which may also threaten genome integrity. AID dynamically shuttles between cytoplasm and nucleus, and the majority stays in the cytoplasm due to active nuclear export mediated by its C-terminal peptide. In immunodeficient-patient cells expressing mutant AID lacking its C-terminus, a catalytically active AID-delC protein accumulates in the nucleus but nevertheless fails to support CSR. To resolve this apparent paradox, we dissected the function of AID-delC proteins in the CSR process and found that they cannot efficiently target antibody genes. We demonstrate that AID-delC proteins form condensates both in vivo and in vitro, dependent on its N-terminus and on a surface arginine-rich patch. Co-expression of AID-delC and wild-type AID leads to an unbalanced nuclear AID-delC/AID ratio, with AID-delC proteins able to trap wild-type AID in condensates, resulting in a dominant-negative phenotype that could contribute to immunodeficiency. The co-condensation model of mutant and wild-type proteins could be an alternative explanation for the dominant-negative effect in genetic disorders.


Asunto(s)
Citidina Desaminasa , Cambio de Clase de Inmunoglobulina , Linfocitos B , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , ADN/metabolismo , Humanos , Cambio de Clase de Inmunoglobulina/genética
10.
Cell ; 144(3): 353-63, 2011 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-21255825

RESUMEN

Activation-induced cytidine deaminase (AID) initiates immunoglobulin (Ig) heavy-chain (IgH) class switch recombination (CSR) and Ig variable region somatic hypermutation (SHM) in B lymphocytes by deaminating cytidines on template and nontemplate strands of transcribed DNA substrates. However, the mechanism of AID access to the template DNA strand, particularly when hybridized to a nascent RNA transcript, has been an enigma. We now implicate the RNA exosome, a cellular RNA-processing/degradation complex, in targeting AID to both DNA strands. In B lineage cells activated for CSR, the RNA exosome associates with AID, accumulates on IgH switch regions in an AID-dependent fashion, and is required for optimal CSR. Moreover, both the cellular RNA exosome complex and a recombinant RNA exosome core complex impart robust AID- and transcription-dependent DNA deamination of both strands of transcribed SHM substrates in vitro. Our findings reveal a role for noncoding RNA surveillance machinery in generating antibody diversity.


Asunto(s)
Linfocitos B/metabolismo , Citidina Desaminasa/metabolismo , Exorribonucleasas/metabolismo , Cambio de Clase de Inmunoglobulina , Cadenas Pesadas de Inmunoglobulina/genética , Complejos Multienzimáticos/metabolismo , ARN/metabolismo , Animales , Linfocitos B/citología , Linfocitos B/enzimología , Línea Celular , Células Cultivadas , Humanos , Ratones , Transcripción Genética
11.
J Biol Chem ; 300(5): 107235, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38552739

RESUMEN

Defects in mitochondrial RNA metabolism have been linked to sensorineural deafness that often occurs as a consequence of damaged or deficient inner ear hair cells. In this report, we investigated the molecular mechanism underlying a deafness-associated tRNAPhe 593T > C mutation that changed a highly conserved uracil to cytosine at position 17 of the DHU-loop. The m.593T > C mutation altered tRNAPhe structure and function, including increased melting temperature, resistance to S1 nuclease-mediated digestion, and conformational changes. The aberrant tRNA metabolism impaired mitochondrial translation, which was especially pronounced by decreases in levels of ND1, ND5, CYTB, CO1, and CO3 harboring higher numbers of phenylalanine. These alterations resulted in aberrant assembly, instability, and reduced activities of respiratory chain enzyme complexes I, III, IV, and intact supercomplexes overall. Furthermore, we found that the m.593T > C mutation caused markedly diminished membrane potential, and increased the production of reactive oxygen species in the mutant cell lines carrying the m.593T > C mutation. These mitochondrial dysfunctions led to the mitochondrial dynamic imbalance via increasing fission with abnormal mitochondrial morphology. Excessive fission impaired the process of autophagy including the initiation phase, formation, and maturation of the autophagosome. In particular, the m.593T > C mutation upregulated the PARKIN-dependent mitophagy pathway. These alterations promoted an intrinsic apoptotic process for the removal of damaged cells. Our findings provide critical insights into the pathophysiology of maternally inherited deafness arising from tRNA mutation-induced defects in mitochondrial and cellular integrity.


Asunto(s)
Sordera , Mitocondrias , ARN de Transferencia de Fenilalanina , Humanos , Autofagia , Sordera/genética , Sordera/metabolismo , Potencial de la Membrana Mitocondrial , Mitocondrias/metabolismo , Mitocondrias/genética , Mitocondrias/patología , Dinámicas Mitocondriales , Mutación , Especies Reactivas de Oxígeno/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , ARN de Transferencia de Fenilalanina/genética
12.
Mol Cell ; 67(3): 361-373.e4, 2017 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-28757211

RESUMEN

Activation-induced cytidine deaminase (AID) initiates both class switch recombination (CSR) and somatic hypermutation (SHM) in antibody diversification. Mechanisms of AID targeting and catalysis remain elusive despite its critical immunological roles and off-target effects in tumorigenesis. Here, we produced active human AID and revealed its preferred recognition and deamination of structured substrates. G-quadruplex (G4)-containing substrates mimicking the mammalian immunoglobulin switch regions are particularly good AID substrates in vitro. By solving crystal structures of maltose binding protein (MBP)-fused AID alone and in complex with deoxycytidine monophosphate, we surprisingly identify a bifurcated substrate-binding surface that explains structured substrate recognition by capturing two adjacent single-stranded overhangs simultaneously. Moreover, G4 substrates induce cooperative AID oligomerization. Structure-based mutations that disrupt bifurcated substrate recognition or oligomerization both compromise CSR in splenic B cells. Collectively, our data implicate intrinsic preference of AID for structured substrates and uncover the importance of G4 recognition and oligomerization of AID in CSR.


Asunto(s)
Citidina Desaminasa/metabolismo , ADN/metabolismo , Cambio de Clase de Inmunoglobulina , Región de Cambio de la Inmunoglobulina , Recombinación Genética , Desaminasas APOBEC/genética , Desaminasas APOBEC/metabolismo , Animales , Diversidad de Anticuerpos , Linfocitos B/enzimología , Linfocitos B/inmunología , Citidina Desaminasa/química , Citidina Desaminasa/genética , ADN/química , ADN/genética , Humanos , Ratones , Modelos Moleculares , Mutación , Conformación de Ácido Nucleico , Unión Proteica , Conformación Proteica , Bazo/enzimología , Bazo/inmunología , Relación Estructura-Actividad , Especificidad por Sustrato
13.
J Immunol ; 208(1): 143-154, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34862258

RESUMEN

Somatic hypermutation (SHM) drives the genetic diversity of Ig genes in activated B cells and supports the generation of Abs with increased affinity for Ag. SHM is targeted to Ig genes by their enhancers (diversification activators [DIVACs]), but how the enhancers mediate this activity is unknown. We show using chicken DT40 B cells that highly active DIVACs increase the phosphorylation of RNA polymerase II (Pol II) and Pol II occupancy in the mutating gene with little or no accompanying increase in elongation-competent Pol II or production of full-length transcripts, indicating accumulation of stalled Pol II. DIVAC has similar effect also in human Ramos Burkitt lymphoma cells. The DIVAC-induced stalling is weakly associated with an increase in the detection of ssDNA bubbles in the mutating target gene. We did not find evidence for antisense transcription, or that DIVAC functions by altering levels of H3K27ac or the histone variant H3.3 in the mutating gene. These findings argue for a connection between Pol II stalling and cis-acting targeting elements in the context of SHM and thus define a mechanistic basis for locus-specific targeting of SHM in the genome. Our results suggest that DIVAC elements render the target gene a suitable platform for AID-mediated mutation without a requirement for increasing transcriptional output.


Asunto(s)
Proteínas Aviares/metabolismo , Subgrupos de Linfocitos B/inmunología , Linfoma de Burkitt/inmunología , Elementos de Facilitación Genéticos/genética , Inmunoglobulinas/metabolismo , ARN Polimerasa II/metabolismo , Animales , Diversidad de Anticuerpos , Proteínas Aviares/genética , Linfoma de Burkitt/genética , Pollos , Citidina Desaminasa/genética , Humanos , Inmunoglobulinas/genética , Activación de Linfocitos , Mutagénesis Sitio-Dirigida , Mutación/genética , ARN Polimerasa II/genética , Hipermutación Somática de Inmunoglobulina , Transcripción Genética
14.
Nucleic Acids Res ; 50(17): 9632-9646, 2022 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-36043443

RESUMEN

Adenine base editors (ABEs) catalyze A-to-G conversions, offering therapeutic options to treat the major class of human pathogenic single nucleotide polymorphisms (SNPs). However, robust and precise editing at diverse genome loci remains challenging. Here, using high-throughput chemical screening, we identified and validated SB505124, a selective ALK5 inhibitor, as an ABE activator. Treating cells with SB505124 enhanced on-target editing at multiple genome loci, including epigenetically refractory regions, and showed little effect on off-target conversion on the genome. Furthermore, SB505124 facilitated the editing of disease-associated genes in vitro and in vivo. Intriguingly, SB505124 served as a specific activator by selectively promoting ABE activity. Mechanistically, SB505124 promotes ABE editing, at least in part, by enhancing ABE expression and modulating DNA repair-associated genes. Our findings reveal the role of the canonical transforming growth factor-ß pathway in gene editing and equip ABEs with precise chemical control.


Asunto(s)
Adenina , Factor de Crecimiento Transformador beta , Adenina/química , Sistemas CRISPR-Cas , Edición Génica , Genoma , Humanos , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factores de Crecimiento Transformadores/metabolismo
15.
Nucleic Acids Res ; 50(16): 9453-9469, 2022 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-36039763

RESUMEN

In this report, we investigated the molecular mechanism underlying a deafness-associated m.5783C > T mutation that affects the canonical C50-G63 base-pairing of TΨC stem of tRNACys and immediately adjacent to 5' end of light-strand origin of mitochondrial DNA (mtDNA) replication (OriL). Two dimensional agarose gel electrophoresis revealed marked decreases in the replication intermediates including ascending arm of Y-fork arcs spanning OriL in the mutant cybrids bearing m.5783C > T mutation. mtDNA replication alterations were further evidenced by decreased levels of PolγA, Twinkle and SSBP1, newly synthesized mtDNA and mtDNA contents in the mutant cybrids. The m.5783C > T mutation altered tRNACys structure and function, including decreased melting temperature, conformational changes, instability and deficient aminoacylation of mutated tRNACys. The m.5783C > T mutation impaired the 5' end processing efficiency of tRNACys precursors and reduced the levels of tRNACys and downstream tRNATyr. The aberrant tRNA metabolism impaired mitochondrial translation, which was especially pronounced effects in the polypeptides harboring higher numbers of cysteine and tyrosine codons. These alterations led to deficient oxidative phosphorylation including instability and reduced activities of the respiratory chain enzyme complexes I, III, IV and intact supercomplexes overall. Our findings highlight the impact of mitochondrial dysfunction on deafness arising from defects in mitochondrial DNA replication and tRNA metabolism.


Asunto(s)
ADN Mitocondrial , Sordera , Humanos , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , ARN de Transferencia de Cisteína/metabolismo , Sordera/genética , Sordera/metabolismo , Mitocondrias/metabolismo , Mutación , Replicación del ADN/genética , Proteínas de Unión al ADN/genética , Proteínas Mitocondriales/metabolismo
16.
Nucleic Acids Res ; 50(16): 9368-9381, 2022 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-36018806

RESUMEN

Pseudouridine (Ψ) at position 55 in tRNAs plays an important role in their structure and function. This modification is catalyzed by TruB/Pus4/Cbf5 family of pseudouridine synthases in bacteria and yeast. However, the mechanism of TRUB family underlying the formation of Ψ55 in the mammalian tRNAs is largely unknown. In this report, the CMC/reverse transcription assays demonstrated the presence of Ψ55 in the human mitochondrial tRNAAsn, tRNAGln, tRNAGlu, tRNAPro, tRNAMet, tRNALeu(UUR) and tRNASer(UCN). TRUB1 knockout (KO) cell lines generated by CRISPR/Cas9 technology exhibited the loss of Ψ55 modification in mitochondrial tRNAAsn, tRNAGln, tRNAGlu and tRNAPro but did not affect other 18 mitochondrial tRNAs. An in vitro assay revealed that recombinant TRUB1 protein can catalyze the efficient formation of Ψ55 in tRNAAsn and tRNAGln, but not in tRNAMet and tRNAArg. Notably, the overexpression of TRUB1 cDNA reversed the deficient Ψ55 modifications in these tRNAs in TRUB1KO HeLa cells. TRUB1 deficiency affected the base-pairing (18A/G-Ψ55), conformation and stability but not aminoacylation capacity of these tRNAs. Furthermore, TRUB1 deficiency impacted mitochondrial translation and biogenesis of oxidative phosphorylation system. Our findings demonstrated that human TRUB1 is a highly conserved mitochondrial pseudouridine synthase responsible for the Ψ55 modification in the mitochondrial tRNAAsn, tRNAGln, tRNAGlu and tRNAPro.


Asunto(s)
Transferasas Intramoleculares , ARN de Transferencia de Ácido Glutámico , Animales , Humanos , ARN de Transferencia de Glutamina , ARN de Transferencia de Prolina , ARN de Transferencia de Asparagina , ARN de Transferencia de Metionina , Células HeLa , Transferasas Intramoleculares/genética , Transferasas Intramoleculares/metabolismo , Seudouridina/genética , Seudouridina/metabolismo , ARN de Transferencia/metabolismo , Mamíferos/genética
17.
Cell Mol Biol (Noisy-le-grand) ; 69(15): 103-107, 2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-38279478

RESUMEN

Colon cancer is one of the most common cancers affecting many people worldwide. This disease can be treated if diagnosed in the early stages. Therefore, with the hypothesis that the level of expression of inflammatory genes in peripheral blood monocytes of patients with colon cancer is different from that of healthy people, this research was done to find out the role of inflammation in the development of colon cancer by relying on its immunopathological profile to help diagnose it in the early stages. In this case-control study, the expression levels of TLR4, TLR2, NLRP3, and NOS2 genes in 15 patients with confirmed stage II colon cancer were determined by the TNM method. Also, 15 healthy people referred for this cancer screening were selected as the control group. First, RNA was extracted from the blood monocytes of two groups, and after making cDNA, the comparison was created using the qPCR method. In this study, the ß-actin gene was used as a reference gene. The expression levels of TLR2 and TLR4 at the mRNA level were significantly lower in colon cancer patients compared to the healthy control group (P<0.05). The expression level of NLRP3 in the group of colon cancer patients showed a relative increase. Still, it was not significant, while the expression level of the NOS2 gene in the group of colon cancer patients increased significantly compared to the healthy control group (P<0.05). Considering the significant changes in TLR4, TLR2, and NOS2 gene expression in monocytes of patients with grade II colon cancer and the role of inflammatory reactions in the development of this cancer, these findings can be used to diagnose and determine the prognosis. However, this requires further studies.


Asunto(s)
Neoplasias del Colon , Monocitos , Humanos , Monocitos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Estudios de Casos y Controles , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Neoplasias del Colon/genética , Expresión Génica
18.
Nature ; 542(7642): 489-493, 2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28199309

RESUMEN

Activation-induced cytidine deaminase (AID) is a B-cell-specific enzyme that targets immunoglobulin genes to initiate class switch recombination and somatic hypermutation. In addition, through off-target activity, AID has a much broader effect on genomic instability by initiating oncogenic chromosomal translocations and mutations involved in the development and progression of lymphoma. AID expression is tightly regulated in B cells and its overexpression leads to enhanced genomic instability and lymphoma formation. The phosphatidylinositol 3-kinase δ (PI3Kδ) pathway regulates AID by suppressing its expression in B cells. Drugs for leukaemia or lymphoma therapy such as idelalisib, duvelisib and ibrutinib block PI3Kδ activity directly or indirectly, potentially affecting AID expression and, consequently, genomic stability in B cells. Here we show that treatment of primary mouse B cells with idelalisib or duvelisib, and to a lesser extent ibrutinib, enhanced the expression of AID and increased somatic hypermutation and chromosomal translocation frequency to the Igh locus and to several AID off-target sites. Both of these effects were completely abrogated in AID-deficient B cells. PI3Kδ inhibitors or ibrutinib increased the formation of AID-dependent tumours in pristane-treated mice. Consistently, PI3Kδ inhibitors enhanced AID expression and translocation frequency to IGH and AID off-target sites in human chronic lymphocytic leukaemia and mantle cell lymphoma cell lines, and patients treated with idelalisib, but not ibrutinib, showed increased somatic hypermutation in AID off-targets. In summary, we show that PI3Kδ or Bruton's tyrosine kinase inhibitors increase genomic instability in normal and neoplastic B cells by an AID-dependent mechanism. This effect should be carefully considered, as such inhibitors can be administered to patients for years.


Asunto(s)
Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Inestabilidad Genómica/efectos de los fármacos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Adenina/análogos & derivados , Agammaglobulinemia Tirosina Quinasa , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Linfocitos B/enzimología , Linfocitos B/patología , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Citidina Desaminasa/metabolismo , Inhibidores Enzimáticos/efectos adversos , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Cambio de Clase de Inmunoglobulina/efectos de los fármacos , Cadenas Pesadas de Inmunoglobulina/genética , Isoquinolinas/efectos adversos , Isoquinolinas/farmacología , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/patología , Linfoma de Células del Manto/genética , Linfoma de Células del Manto/patología , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Piperidinas , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Purinas/efectos adversos , Purinas/farmacología , Pirazoles/efectos adversos , Pirazoles/farmacología , Pirimidinas/efectos adversos , Pirimidinas/farmacología , Quinazolinonas/efectos adversos , Quinazolinonas/farmacología , Recombinación Genética/efectos de los fármacos , Hipermutación Somática de Inmunoglobulina/efectos de los fármacos , Translocación Genética/efectos de los fármacos
19.
Nucleic Acids Res ; 49(15): 8732-8742, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34365511

RESUMEN

CRISPR-Cas9 generates double-stranded DNA breaks (DSBs) to activate cellular DNA repair pathways for genome editing. The repair of DSBs leads to small insertions or deletions (indels) and other complex byproducts, including large deletions and chromosomal translocations. Indels are well understood to disrupt target genes, while the other deleterious byproducts remain elusive. We developed a new in silico analysis pipeline for the previously described primer-extension-mediated sequencing assay to comprehensively characterize CRISPR-Cas9-induced DSB repair outcomes in human or mouse cells. We identified tremendous deleterious DSB repair byproducts of CRISPR-Cas9 editing, including large deletions, vector integrations, and chromosomal translocations. We further elucidated the important roles of microhomology, chromosomal interaction, recurrent DSBs, and DSB repair pathways in the generation of these byproducts. Our findings provide an extra dimension for genome editing safety besides off-targets. And caution should be exercised to avoid not only off-target damages but also deleterious DSB repair byproducts during genome editing.


Asunto(s)
Proteína 9 Asociada a CRISPR , Sistemas CRISPR-Cas , Reparación del ADN , Edición Génica , Animales , Células Cultivadas , Simulación por Computador , Humanos , Ratones , Plásmidos/genética , Eliminación de Secuencia , Translocación Genética
20.
Nucleic Acids Res ; 49(2): 1075-1093, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33398350

RESUMEN

Defects in the posttranscriptional modifications of mitochondrial tRNAs have been linked to human diseases, but their pathophysiology remains elusive. In this report, we investigated the molecular mechanism underlying a deafness-associated tRNAIle 4295A>G mutation affecting a highly conserved adenosine at position 37, 3' adjacent to the tRNA's anticodon. Primer extension and methylation activity assays revealed that the m.4295A>G mutation introduced a tRNA methyltransferase 5 (TRMT5)-catalyzed m1G37 modification of tRNAIle. Molecular dynamics simulations suggested that the m.4295A>G mutation affected tRNAIle structure and function, supported by increased melting temperature, conformational changes and instability of mutated tRNA. An in vitro processing experiment revealed that the m.4295A>G mutation reduced the 5' end processing efficiency of tRNAIle precursors, catalyzed by RNase P. We demonstrated that cybrid cell lines carrying the m.4295A>G mutation exhibited significant alterations in aminoacylation and steady-state levels of tRNAIle. The aberrant tRNA metabolism resulted in the impairment of mitochondrial translation, respiratory deficiency, decreasing membrane potentials and ATP production, increasing production of reactive oxygen species and promoting autophagy. These demonstrated the pleiotropic effects of m.4295A>G mutation on tRNAIle and mitochondrial functions. Our findings highlighted the essential role of deficient posttranscriptional modifications in the structure and function of tRNA and their pathogenic consequence of deafness.


Asunto(s)
Pérdida Auditiva Sensorineural/genética , Mutación Puntual , ARN de Transferencia de Isoleucina/genética , Adenosina Trifosfato/biosíntesis , Adulto , Proteínas Arqueales/metabolismo , Autofagia , Secuencia de Bases , Línea Celular , ADN Mitocondrial/genética , Etnicidad/genética , Femenino , Pleiotropía Genética , Pérdida Auditiva Sensorineural/etnología , Humanos , Isoleucina/metabolismo , Masculino , Herencia Materna , Potencial de la Membrana Mitocondrial , Methanocaldococcus/enzimología , Metilación , Persona de Mediana Edad , Mitocondrias/metabolismo , Simulación de Dinámica Molecular , Conformación de Ácido Nucleico , Fosforilación Oxidativa , Linaje , Biosíntesis de Proteínas , Procesamiento Postranscripcional del ARN , Proteínas Recombinantes/metabolismo , Aminoacilación de ARN de Transferencia , Adulto Joven , ARNt Metiltransferasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA