Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 19(2): e1011156, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36745676

RESUMEN

Human adenoviruses (HAdVs) are a large family of DNA viruses counting more than a hundred strains divided into seven species (A to G). HAdVs induce respiratory tract infections, gastroenteritis and conjunctivitis. APOBEC3B is a cytidine deaminase that restricts several DNA viruses. APOBEC3B is also implicated in numerous cancers where it is responsible for the introduction of clustered mutations into the cellular genome. In this study, we demonstrate that APOBEC3B is an adenovirus restriction factor acting through a deaminase-dependent mechanism. APOBEC3B introduces C-to-T clustered mutations into the adenovirus genome. APOBEC3B reduces the propagation of adenoviruses by limiting viral genome replication, progression to late phase, and production of infectious virions. APOBEC3B restriction efficiency varies between adenoviral strains, the A12 strain being more sensitive to APOBEC3B than the B3 or C2 strains. In A12-infected cells, APOBEC3B clusters in the viral replication centers. Importantly, we show that adenovirus infection leads to a reduction of the quantity and/or enzymatic activity of the APOBEC3B protein depending on the strains. The A12 strain seems less able to resist APOBEC3B than the B3 or C2 strains, a characteristic which could explain the strong depletion of the APOBEC3-targeted motifs in the A12 genome. These findings suggest that adenoviruses evolved different mechanisms to antagonize APOBEC3B. Elucidating these mechanisms could benefit the design of cancer treatments. This study also identifies adenoviruses as triggers of the APOBEC3B-mediated innate response. The involvement of certain adenoviral strains in the genesis of the APOBEC3 mutational signature observed in tumors deserves further study.


Asunto(s)
Infecciones por Adenoviridae , Neoplasias , Humanos , Adenoviridae/genética , Adenoviridae/metabolismo , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Proteínas/metabolismo , Neoplasias/patología , Antígenos de Histocompatibilidad Menor/genética
2.
PLoS Pathog ; 19(2): e1011170, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36802406

RESUMEN

Viruses have evolved countless mechanisms to subvert and impair the host innate immune response. Measles virus (MeV), an enveloped, non-segmented, negative-strand RNA virus, alters the interferon response through different mechanisms, yet no viral protein has been described as directly targeting mitochondria. Among the crucial mitochondrial enzymes, 5'-aminolevulinate synthase (ALAS) is an enzyme that catalyzes the first step in heme biosynthesis, generating 5'-aminolevulinate from glycine and succinyl-CoA. In this work, we demonstrate that MeV impairs the mitochondrial network through the V protein, which antagonizes the mitochondrial enzyme ALAS1 and sequesters it to the cytosol. This re-localization of ALAS1 leads to a decrease in mitochondrial volume and impairment of its metabolic potential, a phenomenon not observed in MeV deficient for the V gene. This perturbation of the mitochondrial dynamics demonstrated both in culture and in infected IFNAR-/- hCD46 transgenic mice, causes the release of mitochondrial double-stranded DNA (mtDNA) in the cytosol. By performing subcellular fractionation post infection, we demonstrate that the most significant source of DNA in the cytosol is of mitochondrial origin. Released mtDNA is then recognized and transcribed by the DNA-dependent RNA polymerase III. The resulting double-stranded RNA intermediates will be captured by RIG-I, ultimately initiating type I interferon production. Deep sequencing analysis of cytosolic mtDNA editing divulged an APOBEC3A signature, primarily analyzed in the 5'TpCpG context. Finally, in a negative feedback loop, APOBEC3A an interferon inducible enzyme will orchestrate the catabolism of mitochondrial DNA, decrease cellular inflammation, and dampen the innate immune response.


Asunto(s)
Interferones , Mitocondrias , Ratones , Animales , Mitocondrias/metabolismo , Virus del Sarampión , 5-Aminolevulinato Sintetasa/genética , 5-Aminolevulinato Sintetasa/metabolismo , ADN Mitocondrial
3.
J Infect Dis ; 228(10): 1421-1429, 2023 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-37224627

RESUMEN

BACKGROUND: On May 6, 2022, a powerful outbreak of monkeypox virus (MPXV) had been reported outside of Africa, with many continuing new cases being reported around the world. Analysis of mutations among the 2 different lineages present in the 2021 and 2022 outbreaks revealed the presence of G->A mutations occurring in the 5'GpA context, indicative of APOBEC3 cytidine deaminase activity. METHODS: By using a sensitive polymerase chain reaction (differential DNA denaturation PCR) method allowing differential amplification of AT-rich DNA, we analyzed the level of APOBEC3-induced MPXV editing in infected cells and in patients. RESULTS: We demonstrate that G->A hypermutated MPXV genomes can be recovered experimentally from APOBEC3 transfection followed by MPXV infection. Here, among the 7 human APOBEC3 cytidine deaminases (A3A-A3C, A3DE, A3F-A3H), only APOBEC3F was capable of extensively deaminating cytidine residues in MPXV genomes. Hyperedited genomes were also recovered in ∼42% of analyzed patients. Moreover, we demonstrate that substantial repair of these mutations occurs. Upon selection, corrected G->A mutations escaping drift loss contribute to the MPXV evolution observed in the current epidemic. CONCLUSIONS: Stochastic or transient overexpression of the APOBEC3F gene exposes the MPXV genome to a broad spectrum of mutations that may be modeling the mutational landscape after multiple cycles of viral replication.


Asunto(s)
Citidina Desaminasa , Monkeypox virus , Humanos , Monkeypox virus/genética , Citidina Desaminasa/genética , Mutación , Brotes de Enfermedades , Citidina , Citosina Desaminasa/química , Citosina Desaminasa/genética
4.
J Infect Dis ; 226(5): 891-895, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-35022749

RESUMEN

Single-nucleotide polymorphism in APOBEC3C (resulting in a serine to isoleucine in position 188) is present in approximately 10% of African populations and greatly enhances restriction against human immunodeficiency virus-1 and simian immunodeficiency virus by improving dimerization and DNA processivity of the enzyme. In this study, we demonstrated in culture and in infected patients that hepatitis B virus (HBV) could be edited by APOBEC3CS188I. Using next-generation sequencing, we demonstrated that APOBEC3CS188I led to enhanced editing activity in 5'TpCpA→5'TpTpA context. This constitutes a new hallmark of this enzyme, which could be used to determine its impact on HBV or nuclear DNA.


Asunto(s)
Citidina Desaminasa , Genoma Viral , Virus de la Hepatitis B , Citidina Desaminasa/genética , Hepatitis B/genética , Virus de la Hepatitis B/genética , Humanos , Polimorfismo de Nucleótido Simple
5.
J Biol Chem ; 297(3): 101081, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34403699

RESUMEN

The human APOBEC3A (A3A) cytidine deaminase is a powerful DNA mutator enzyme recognized as a major source of somatic mutations in tumor cell genomes. However, there is a discrepancy between APOBEC3A mRNA levels after interferon stimulation in myeloid cells and A3A detection at the protein level. To understand this difference, we investigated the expression of two novel alternative "A3Alt" proteins encoded in the +1-shifted reading frame of the APOBEC3A gene. A3Alt-L and its shorter isoform A3Alt-S appear to be transmembrane proteins targeted to the mitochondrial compartment that induce membrane depolarization and apoptosis. Thus, the APOBEC3A gene represents a new example wherein a single gene encodes two proapoptotic proteins, A3A cytidine deaminases that target the genome and A3Alt proteins that target mitochondria.


Asunto(s)
Citidina Desaminasa/genética , Citidina Desaminasa/fisiología , Mitocondrias/genética , Proteínas/genética , Proteínas/fisiología , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Citidina Desaminasa/metabolismo , ADN/genética , Mutación del Sistema de Lectura/genética , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Genoma/genética , Humanos , Mitocondrias/metabolismo , Mutación/genética , Proteínas/metabolismo , ARN Mensajero/genética , Sistemas de Lectura/genética
6.
BMC Genomics ; 20(1): 858, 2019 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-31726973

RESUMEN

BACKGROUND: APOBEC1 (A1) enzymes are cytidine deaminases involved in RNA editing. In addition to this activity, a few A1 enzymes have been shown to be active on single stranded DNA. As two human ssDNA cytidine deaminases APOBEC3A (A3A), APOBEC3B (A3B) and related enzymes across the spectrum of placental mammals have been shown to introduce somatic mutations into nuclear DNA of cancer genomes, we explored the mutagenic threat of A1 cytidine deaminases to chromosomal DNA. RESULTS: Molecular cloning and expression of various A1 enzymes reveal that the cow, pig, dog, rabbit and mouse A1 have an intracellular ssDNA substrate specificity. However, among all the enzymes studied, mouse A1 appears to be singular, being able to introduce somatic mutations into nuclear DNA with a clear 5'TpC editing context, and to deaminate 5-methylcytidine substituted DNA which are characteristic features of the cancer related mammalian A3A and A3B enzymes. However, mouse A1 activity fails to elicit formation of double stranded DNA breaks, suggesting that mouse A1 possess an attenuated nuclear DNA mutator phenotype reminiscent of human A3B. CONCLUSIONS: At an experimental level mouse APOBEC1 is remarkable among 12 mammalian A1 enzymes in that it represents a source of somatic mutations in mouse genome, potentially fueling oncogenesis. While the order Rodentia is bereft of A3A and A3B like enzymes it seems that APOBEC1 may well substitute for it, albeit remaining much less active. This modifies the paradigm that APOBEC3 and AID enzymes are the sole endogenous mutator enzymes giving rise to off-target editing of mammalian genomes.


Asunto(s)
Desaminasas APOBEC-1/metabolismo , Cromosomas de los Mamíferos/genética , Mutación , Desaminasas APOBEC-1/química , Desaminasas APOBEC-1/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Roturas del ADN de Doble Cadena , ADN de Cadena Simple , Activación Enzimática , Expresión Génica , Ratones , Filogenia , Edición de ARN , Especificidad por Sustrato
7.
Nucleic Acids Res ; 45(6): 3231-3241, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28100701

RESUMEN

Foreign and self-cytoplasmic DNA are recognized by numerous DNA sensor molecules leading to the production of type I interferons. Such DNA agonists should be degraded otherwise cells would be chronically stressed. Most human APOBEC3 cytidine deaminases can initiate catabolism of cytoplasmic mitochondrial DNA. Using the human myeloid cell line THP-1 with an interferon inducible APOBEC3A gene, we show that cytoplasmic DNA triggers interferon α and ß production through the RNA polymerase III transcription/RIG-I pathway leading to massive upregulation of APOBEC3A. By catalyzing C→U editing in single stranded DNA fragments, the enzyme prevents them from re-annealing so attenuating the danger signal. The price to pay is chromosomal DNA damage in the form of CG→TA mutations and double stranded DNA breaks which, in the context of chronic inflammation, could drive cells down the path toward cancer.


Asunto(s)
Citidina Desaminasa/biosíntesis , Roturas del ADN de Doble Cadena , ADN Mitocondrial/metabolismo , Línea Celular Tumoral , Cromosomas Humanos , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Citosol/metabolismo , Proteína 58 DEAD Box , ADN Mitocondrial/química , Humanos , Interferón-alfa/biosíntesis , Interferón beta/biosíntesis , Interferón beta/fisiología , Proteínas/genética , Proteínas/metabolismo , ARN Polimerasa III/metabolismo , Receptores Inmunológicos , Transcripción Genética , Regulación hacia Arriba , Uracilo/metabolismo
8.
Nucleic Acids Res ; 43(19): 9340-9, 2015 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-26384561

RESUMEN

The human APOBEC3A and APOBEC3B genes (A3A and A3B) encode DNA mutator enzymes that deaminate cytidine and 5-methylcytidine residues in single-stranded DNA (ssDNA). They are important sources of mutations in many cancer genomes which show a preponderance of CG->TA transitions. Although both enzymes can hypermutate chromosomal DNA in an experimental setting, only A3A can induce double strand DNA breaks, even though the catalytic domains of A3B and A3A differ by only 9% at the protein level. Accordingly we sought the molecular basis underlying A3B attenuation through the generation of A3A-A3B chimeras and mutants. It transpires that the N-terminal domain facilitates A3B activity while a handful of substitutions in the catalytic C-terminal domain impacting ssDNA binding serve to attenuate A3B compared to A3A. Interestingly, functional attenuation is also observed for the rhesus monkey rhA3B enzyme compared to rhA3A indicating that this genotoxic dichotomy has been selected for and maintained for some 38 million years. Expression of all human ssDNA cytidine deaminase genes is absent in mature sperm indicating they contribute to somatic mutation and cancer but not human diversity.


Asunto(s)
Citidina Desaminasa/genética , Roturas del ADN de Doble Cadena , Animales , Línea Celular , Citidina Desaminasa/química , Citidina Desaminasa/metabolismo , Células HeLa , Humanos , Macaca mulatta , Antígenos de Histocompatibilidad Menor , Mutación , Fenotipo , Estructura Terciaria de Proteína , Proteínas/química , Proteínas/genética , Codorniz , Edición de ARN
9.
Retrovirology ; 13(1): 84, 2016 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-27998270

RESUMEN

BACKGROUND: The replication of HBV involves the production of covalently closed circular DNA (cccDNA) from the HBV genome through the repair of virion relaxed circular DNA (rcDNA) in the virion. As cccDNA is the transcription template for HBV genomes, it needs to be eliminated from hepatocytes if the eradication of chronic HBV infection is to be achieved. PCR quantitation of cccDNA copy number is the technique of choice for evaluating the efficiency of treatment regimens. The PCR target commonly used to identify cccDNA spans the gapped region of rcDNA and is considered to accurately distinguish between cccDNA and rcDNA. There is however, a potentially confounding issue in that PCR can generate larger targets from collections of small DNA fragments, a phenomenon known as PCR recombination. RESULTS: The impact of PCR recombination towards the amplification of this cccDNA specific target was explored by mixing three marked, yet overlapping HBV DNA fragments. Thirteen of sixteen possible recombinants were identified by sequencing with frequencies ranging from 0.6 to 23%. To confirm this finding in vivo, HBV positive sera were treated with DNase I and submitted to quantitative real-time PCR. Under these conditions, it was possible to amplify the cccDNA specific segment without difficulty. As the virion contains uniquely rcDNA, amplification of the cccDNA target resulted from PCR recombination. CONCLUSIONS: PCR quantitation of cccDNA may be more difficult than hitherto thought. Current detection protocols need to be investigated so as to help in the management of chronic HBV infection.


Asunto(s)
ADN Circular/análisis , ADN Viral/análisis , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/aislamiento & purificación , Hepatitis B/virología , Reacción en Cadena en Tiempo Real de la Polimerasa , ADN Circular/sangre , ADN Viral/sangre , ADN Viral/genética , Virus de la Hepatitis B/fisiología , Hepatitis B Crónica/virología , Humanos , Virión/genética , Replicación Viral
10.
Mol Biol Evol ; 31(2): 330-40, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24162735

RESUMEN

The human APOBEC3 gene cluster locus encodes polynucleotide cytidine deaminases. Although many act as viral restriction factors through mutation of single-stranded DNA, recent reports have shown that human APOBEC3A was capable of efficiently hypermutating nuclear DNA and inducing DNA breaks in genomic DNA. In addition, the enzyme was unique in efficiently deaminating 5-methylcytidine in single-stranded DNA. To appreciate the evolutionary relevance of these activities, we analyzed A3A-related enzymes from the rhesus and tamarin monkey, horse, sheep, dog, and panda. All proved to be orthologous to the human enzyme in all these activities revealing strong conservation more than 148 My. Hence, their singular role in DNA catabolism is a well-established mechanism probably outweighing any deleterious or pathological roles such as genomic instability and cancer formation.


Asunto(s)
Citidina Desaminasa/metabolismo , Citidina/análogos & derivados , ADN/genética , Mamíferos/metabolismo , Secuencia de Aminoácidos , Animales , Citidina/metabolismo , Citidina Desaminasa/química , Citidina Desaminasa/genética , Roturas del ADN de Doble Cadena , Desaminación , Perros , Inestabilidad Genómica , Células HEK293 , Células HeLa , Humanos , Células de Riñón Canino Madin Darby , Mamíferos/genética , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Filogenia , Estructura Secundaria de Proteína , Proteínas/química , Proteínas/genética , Proteínas/metabolismo , Alineación de Secuencia
11.
Retrovirology ; 11: 93, 2014 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-25389016

RESUMEN

BACKGROUND: The role of innate immunity in general and of type I interferon (IFN-I) in particular in HTLV-1 pathogenesis is still a matter of debate. ADAR1-p150 is an Interferon Stimulated Gene (ISG) induced by IFN-I that can edit viral RNAs. We therefore investigated whether it could play the role of an anti-HTLV factor. RESULTS: We demonstrate here that ADAR1 is also expressed in the absence of IFN stimulation in activated primary T-lymphocytes that are the natural target of this virus and in HTLV-1 or HTLV-2 chronically infected T-cells. ADAR1 expression is also increased in primary lymphocytes obtained from HTLV-1 infected individuals. We show that ADAR1 enhances HTLV-1 and HTLV-2 infection in T-lymphocytes and that this proviral effect is independent from its editing activity. ADAR1 expression suppresses IFN-α inhibitory effect on HTLV-1 and HTLV-2 and acts through the repression of PKR phosphorylation. DISCUSSION: This study demonstrates that two interferon stimulated genes, i.e. PKR and ADAR1 have opposite effects on HTLV replication in vivo. The balanced expression of those proteins could determine the fate of the viral cycle in the course of infection.


Asunto(s)
Adenosina Desaminasa/metabolismo , Interacciones Huésped-Patógeno , Virus Linfotrópico T Tipo 1 Humano/fisiología , Virus Linfotrópico T Tipo 2 Humano/fisiología , Proteínas de Unión al ARN/metabolismo , Replicación Viral , eIF-2 Quinasa/antagonistas & inhibidores , Células Cultivadas , Humanos , Inhibición Psicológica , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Linfocitos T/inmunología , Linfocitos T/virología
12.
Proc Natl Acad Sci U S A ; 108(12): 4858-63, 2011 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-21368204

RESUMEN

The human APOBEC3 (A3A-A3H) locus encodes six cytidine deaminases that edit single-stranded DNA, the result being DNA peppered with uridine. Although several cytidine deaminases are clearly restriction factors for retroviruses and hepadnaviruses, it is not known if APOBEC3 enzymes have roles outside of these settings. It is shown here that both human mitochondrial and nuclear DNA are vulnerable to somatic hypermutation by A3 deaminases, with APOBEC3A standing out among them. The degree of editing is much greater in patients lacking the uracil DNA-glycolyase gene, indicating that the observed levels of editing reflect a dynamic composed of A3 editing and DNA catabolism involving uracil DNA-glycolyase. Nonetheless, hyper- and lightly mutated sequences went hand in hand, raising the hypothesis that recurrent low-level mutation by APOBEC3A could catalyze the transition from a healthy to a cancer genome.


Asunto(s)
Citosina Desaminasa/metabolismo , ADN Mitocondrial/metabolismo , Sitios Genéticos , Genoma Humano , Mutación , Desaminasas APOBEC , Citidina Desaminasa , Citosina Desaminasa/genética , ADN Mitocondrial/genética , Femenino , Células HeLa , Hepadnaviridae/genética , Hepadnaviridae/metabolismo , Humanos , Masculino , Neoplasias/enzimología , Neoplasias/genética , Retroviridae/genética , Retroviridae/metabolismo , Uracil-ADN Glicosidasa/deficiencia , Uracil-ADN Glicosidasa/genética , Uracil-ADN Glicosidasa/metabolismo
13.
Stem Cell Res Ther ; 15(1): 10, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-38167524

RESUMEN

BACKGROUND: Beyond the observed alterations in cellular structure and mitochondria, the mechanisms linking rare genetic mutations to the development of heart failure in patients affected by desmin mutations remain unclear due in part, to the lack of relevant human cardiomyocyte models. METHODS: To shed light on the role of mitochondria in these mechanisms, we investigated cardiomyocytes derived from human induced pluripotent stem cells carrying the heterozygous DESE439K mutation that were either isolated from a patient or generated by gene editing. To increase physiological relevance, cardiomyocytes were either cultured on an anisotropic micropatterned surface to obtain elongated and aligned cardiomyocytes, or as a cardiac spheroid to create a micro-tissue. Moreover, when applicable, results from cardiomyocytes were confirmed with heart biopsies of suddenly died patient of the same family harboring DESE439K mutation, and post-mortem heart samples from five control healthy donors. RESULTS: The heterozygous DESE439K mutation leads to dramatic changes in the overall cytoarchitecture of cardiomyocytes, including cell size and morphology. Most importantly, mutant cardiomyocytes display altered mitochondrial architecture, mitochondrial respiratory capacity and metabolic activity reminiscent of defects observed in patient's heart tissue. Finally, to challenge the pathological mechanism, we transferred normal mitochondria inside the mutant cardiomyocytes and demonstrated that this treatment was able to restore mitochondrial and contractile functions of cardiomyocytes. CONCLUSIONS: This work highlights the deleterious effects of DESE439K mutation, demonstrates the crucial role of mitochondrial abnormalities in the pathophysiology of desmin-related cardiomyopathy, and opens up new potential therapeutic perspectives for this disease.


Asunto(s)
Cardiomiopatías , Células Madre Pluripotentes Inducidas , Humanos , Desmina/genética , Desmina/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Cardiomiopatías/metabolismo , Mutación/genética , Miocitos Cardíacos/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo
14.
J Biol Chem ; 287(46): 39182-92, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-22977230

RESUMEN

The human polydeoxynucleotide cytidine deaminases APOBEC3A, APOBEC3C, and APOBEC3H are capable of mutating viral DNA in the nucleus, whereas APOBEC3A alone efficiently edits nuclear DNA. Deamination is rapidly followed by excision of uracil residues and can lead to double-stranded breaks. It is not known to which protein networks these DNA mutators belong. Using a yeast two-hybrid screen, we identified the human homolog of Drosophila Tribbles 3, TRIB3, as an interactor for APOBEC3A and APOBEC3C. The interaction was confirmed by co-affinity purification. Co-transfection of APOBEC3A with a TRIB3 expression vector reduced nuclear DNA editing whereas siRNA knockdown of TRIB3 increased the levels of nuclear DNA editing, indicating that TRIB3 functioned as a repressor of A3A. It also repressed A3A-associated γH2AX positive double-stranded breaks. The interaction results in degradation of A3A in a proteasome-independent manner. TRIB3 has been linked to cancer and via its own interactors and links the A3A DNA mutators to the Rb-BRCA1-ATM network. TRIB3 emerges as an important guardian of genome integrity.


Asunto(s)
Núcleo Celular/metabolismo , Citidina Desaminasa/metabolismo , Citidina/química , ADN/metabolismo , Regulación de la Expresión Génica , Proteínas/metabolismo , Ciclo Celular , Supervivencia Celular , Citidina Desaminasa/fisiología , Reparación del ADN , Desaminación , Genoma , Células HEK293 , Células HeLa , Humanos , Microscopía Confocal/métodos , Mutagénesis , Mutación , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas/fisiología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Técnicas del Sistema de Dos Híbridos
15.
J Gen Virol ; 93(Pt 12): 2646-2651, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22993189

RESUMEN

RNA editing mediated by adenosine deaminases acting on RNA (ADARs) converts adenosine (A) to inosine (I) residues in dsRNA templates. While ADAR-1-mediated editing was essentially described for RNA viruses, the present work addresses the issue for two δ-retroviruses, human T-cell leukemia virus type 2 and simian T-cell leukemia virus type 3 (HTLV-2 and STLV-3). We examined whether ADAR-1 could edit HTLV-2 and STLV-3 virus genomes in cell culture and in vivo. Using a highly sensitive PCR-based method, referred to as 3DI-PCR, we showed that ADAR-1 could hypermutate adenosine residues in HTLV-2. STLV-3 hypermutation was obtained without using 3DI-PCR, suggesting a higher mutation frequency for this virus. Detailed analysis of the dinucleotide editing context showed preferences for 5' ArA and 5' UrA. In conclusion, the present observations demonstrate that ADAR-1 massively edits HTLV-2 and STLV-3 retroviruses in vitro, but probably remains a rare phenomenon in vivo.


Asunto(s)
Adenosina Desaminasa/metabolismo , Virus Linfotrópico T Tipo 2 Humano/genética , Virus Linfotrópico T Tipo 2 Humano/metabolismo , Edición de ARN/fisiología , ARN Viral/genética , ARN Viral/metabolismo , Virus Linfotrópico T Tipo 3 de los Simios/genética , Virus Linfotrópico T Tipo 3 de los Simios/metabolismo , Adenosina/química , Adenosina Desaminasa/genética , Animales , Secuencia de Bases , Genoma Viral , Células HEK293 , Humanos , Datos de Secuencia Molecular , Mutación , Reacción en Cadena de la Polimerasa/métodos , ARN Viral/química , Proteínas de Unión al ARN , Homología de Secuencia de Ácido Nucleico
16.
J Virol ; 85(23): 12227-40, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21917974

RESUMEN

Lethal mutagenesis or virus transition into error catastrophe is an antiviral strategy that aims at extinguishing a virus by increasing the viral mutation rates during replication. The molecular basis of lethal mutagenesis is largely unknown. Previous studies showed that a critical substitution in the foot-and-mouth disease virus (FMDV) polymerase was sufficient to allow the virus to escape extinction through modulation of the transition types induced by the purine nucleoside analogue ribavirin. This substitution was not detected in mutant spectra of FMDV populations that had not replicated in the presence of ribavirin, using standard molecular cloning and nucleotide sequencing. Here we selectively amplify and analyze low-melting-temperature cDNA duplexes copied from FMDV genome populations passaged in the absence or presence of ribovirin Hypermutated genomes with high frequencies of A and U were present in both ribavirin -treated and untreated populations, but the major effect of ribavirin mutagenesis was to accelerate the occurrence of AU-rich mutant clouds during the early replication rounds of the virus. The standard FMDV quasispecies passaged in the absence of ribavirin included the salient transition-modulating, ribavirin resistance mutation, whose frequency increased in populations treated with ribavirin. Thus, even nonmutagenized FMDV quasispecies include a deep, mutationally biased portion of sequence space, in support of the view that the virus replicates close to the error threshold for maintenance of genetic information.


Asunto(s)
Antivirales/farmacología , Virus de la Fiebre Aftosa/genética , Genes Letales , Genoma Viral , Mutagénesis , Mutación/genética , Ribavirina/farmacología , Animales , Células Cultivadas , Cricetinae , Replicación del ADN , Farmacorresistencia Viral , Virus de la Fiebre Aftosa/efectos de los fármacos , Virus de la Fiebre Aftosa/crecimiento & desarrollo , Riñón/citología , Tasa de Mutación , ARN Mensajero/genética , ARN Viral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Replicación Viral
17.
J Virol ; 85(15): 7594-602, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21632763

RESUMEN

Human APOBEC3 cytidine deaminases target and edit single-stranded DNA, which can be of viral, mitochondrial, or nuclear origin. Retrovirus genomes, such as human immunodeficiency virus (HIV) genomes deficient in the vif gene and the hepatitis B virus genome, are particularly vulnerable. The genomes of some DNA viruses, such as human papillomaviruses, can be edited in vivo and in transfection experiments. Accordingly, herpesviruses should be no exception. This is indeed the case for herpes simplex virus 1 (HSV-1) in tissue culture, where APOBEC3C (A3C) overexpression can reduce virus titers and the particle/PFU ratio ∼10-fold. Nonetheless, A3A, A3G, and AICDA can edit what is presumably a small fraction of HSV genomes in an experimental setting without seriously impacting the viral titer. Hyperediting was found in HSV genomes recovered from 4/8 uncultured buccal lesions. The phenomenon is not restricted to HSV, since hyperedited Epstein-Barr virus (EBV) genomes were readily recovered from 4/5 established cell lines, indicating that episomes are vulnerable to editing. These findings suggest that the widely expressed A3C cytidine deaminase can function as a restriction factor for some human herpesviruses. That the A3C gene is not induced by type I interferons begs the question whether some herpesviruses encode A3C antagonists.


Asunto(s)
Citosina Desaminasa/metabolismo , Genoma Viral , Herpesvirus Humano 1/genética , Herpesvirus Humano 4/genética , Desaminasas APOBEC , Animales , Secuencia de Bases , Chlorocebus aethiops , Citidina Desaminasa , Citosina Desaminasa/genética , ADN/genética , Células HeLa , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 4/fisiología , Humanos , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Homología de Secuencia de Ácido Nucleico , Células Vero , Replicación Viral
18.
J Virol ; 85(5): 2458-62, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21159878

RESUMEN

We sought to examine ADAR-1 editing of measles and influenza virus genomes derived from inactivated seasonal influenza and live attenuated measles virus vaccines grown on chicken cells as the culture substrate. Using highly sensitive 3DI-PCR (R. Suspène et al., Nucleic Acids Res. 36:e72, 2008), it was possible to show that ADAR-1 could hyperdeaminate adenosine residues in both measles virus and influenza virus A genomes. Detailed analysis of the dinucleotide editing context showed preferences for 5'ArA and 5'UrA, which is typical of editing in mammalian cells. The hyperedited mutant frequency, including genomes and antigenomes, was a log greater for influenza virus compared to measles virus, suggesting a greater sensitivity to restriction by ADAR-1.


Asunto(s)
Adenosina Desaminasa/metabolismo , Genoma Viral , Virus de la Influenza A/genética , Gripe Humana/enzimología , Virus del Sarampión/genética , Sarampión/enzimología , Mutación , Adenosina Desaminasa/genética , Animales , Secuencia de Bases , Línea Celular , Chlorocebus aethiops , Humanos , Vacunas contra la Influenza/genética , Gripe Humana/virología , Sarampión/virología , Vacuna Antisarampión/genética , Datos de Secuencia Molecular , Proteínas de Unión al ARN , Estaciones del Año , Vacunas Atenuadas/genética , Células Vero
19.
PLoS Pathog ; 6(5): e1000928, 2010 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-20523896

RESUMEN

DNA viruses, retroviruses and hepadnaviruses, such as hepatitis B virus (HBV), are vulnerable to genetic editing of single stranded DNA by host cell APOBEC3 (A3) cytidine deaminases. At least three A3 genes are up regulated by interferon-alpha in human hepatocytes while ectopic expression of activation induced deaminase (AICDA), an A3 paralog, has been noted in a variety of chronic inflammatory syndromes including hepatitis C virus infection. Yet virtually all studies of HBV editing have confined themselves to analyses of virions from culture supernatants or serum where the frequency of edited genomes is generally low (< or = 10(-2)). We decided to look at the nature and frequency of HBV editing in cirrhotic samples taken during removal of a primary hepatocellular carcinoma. Forty-one cirrhotic tissue samples (10 alcoholic, 10 HBV(+), 11 HBV(+)HCV(+) and 10 HCV(+)) as well as 4 normal livers were studied. Compared to normal liver, 5/7 APOBEC3 genes were significantly up regulated in the order: HCV+/-HBV>HBV>alcoholic cirrhosis. A3C and A3D were up regulated for all groups while the interferon inducible A3G was over expressed in virus associated cirrhosis, as was AICDA in approximately 50% of these HBV/HCV samples. While AICDA can indeed edit HBV DNA ex vivo, A3G is the dominant deaminase in vivo with up to 35% of HBV genomes being edited. Despite these highly deleterious mutant spectra, a small fraction of genomes survive and contribute to loss of HBeAg antigenemia and possibly HBsAg immune escape. In conclusion, the cytokine storm associated with chronic inflammatory responses to HBV and HCV clearly up regulates a number of A3 genes with A3G clearly being a major restriction factor for HBV. Although the mutant spectrum resulting from A3 editing is highly deleterious, a very small part, notably the lightly edited genomes, might help the virus evolve and even escape immune responses.


Asunto(s)
Citidina Desaminasa/genética , ADN Viral/genética , Virus de la Hepatitis B/genética , Hepatitis B/inmunología , Hepatitis B/virología , Desaminasa APOBEC-3G , Anciano , Citidina Desaminasa/metabolismo , ADN de Cadena Simple/genética , Evolución Molecular , Femenino , Regulación Viral de la Expresión Génica/inmunología , Genoma Viral , Antígenos del Núcleo de la Hepatitis B/genética , Vacunas contra Hepatitis B/genética , Vacunas contra Hepatitis B/inmunología , Virus de la Hepatitis B/inmunología , Humanos , Cirrosis Hepática/virología , Masculino , Persona de Mediana Edad , Mutación , Regulación hacia Arriba/fisiología
20.
J Virol ; 84(14): 7278-87, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20463074

RESUMEN

Human T-cell leukemia virus type 1 (HTLV-1) induces cell proliferation after infection, leading to efficient transmission by cell-to-cell contact. After a long latent period, a fraction of carriers develop adult T-cell leukemia (ATL). Genetic changes in the tax gene in ATL cells were reported in about 10% of ATL cases. To determine genetic changes that may occur throughout the provirus, we determined the entire sequence of the HTLV-1 provirus in 60 ATL cases. Abortive genetic changes, including deletions, insertions, and nonsense mutations, were frequent in all viral genes except the HBZ gene, which is transcribed from the minus strand of the virus. G-to-A base substitutions were the most frequent mutations in ATL cells. The sequence context of G-to-A mutations was in accordance with the preferred target sequence of human APOBEC3G (hA3G). The target sequences of hA3G were less frequent in the plus strand of the HBZ coding region than in other coding regions of the HTLV-1 provirus. Nonsense mutations in viral genes including tax were also observed in proviruses from asymptomatic carriers, indicating that these mutations were generated during reverse transcription and prior to oncogenesis. The fact that hA3G targets the minus strand during reverse transcription explains why the HBZ gene is not susceptible to such nonsense mutations. HTLV-1-infected cells likely take advantage of hA3G to escape from the host immune system by losing expression of viral proteins.


Asunto(s)
Citidina Desaminasa/metabolismo , Genoma Viral , Infecciones por HTLV-I/virología , Virus Linfotrópico T Tipo 1 Humano/genética , Leucemia-Linfoma de Células T del Adulto/virología , Mutación , Provirus/genética , Desaminasa APOBEC-3G , Secuencia de Bases , Línea Celular , Citidina Desaminasa/genética , Genes Reporteros , Variación Genética , Vectores Genéticos , Virus Linfotrópico T Tipo 1 Humano/inmunología , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Humanos , Datos de Secuencia Molecular , Mutagénesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA