Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 297(3): 101081, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34403699

RESUMO

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.


Assuntos
Citidina Desaminase/genética , Citidina Desaminase/fisiologia , Mitocôndrias/genética , Proteínas/genética , Proteínas/fisiologia , Apoptose/genética , Proteínas Reguladoras de Apoptose/genética , Citidina Desaminase/metabolismo , DNA/genética , Mutação da Fase de Leitura/genética , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Genoma/genética , Humanos , Mitocôndrias/metabolismo , Mutação/genética , Proteínas/metabolismo , RNA Mensageiro/genética , Fases de Leitura/genética
2.
BMC Genomics ; 20(1): 858, 2019 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-31726973

RESUMO

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.


Assuntos
Desaminase APOBEC-1/metabolismo , Cromossomos de Mamíferos/genética , Mutação , Desaminase APOBEC-1/química , Desaminase APOBEC-1/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Quebras de DNA de Cadeia Dupla , DNA de Cadeia Simples , Ativação Enzimática , Expressão Gênica , Camundongos , Filogenia , Edição de RNA , Especificidade por Substrato
3.
Cancer Immunol Immunother ; 68(4): 533-544, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30656384

RESUMO

Cancer immunotherapy is seeing an increasing focus on vaccination with tumor-associated antigens (TAAs). Human telomerase (hTERT) is a TAA expressed by most tumors to overcome telomere shortening. Tolerance to hTERT can be easily broken both naturally and experimentally and hTERT DNA vaccine candidates have been introduced in clinical trials. DNA prime/boost strategies have been widely developed to immunize efficiently against infectious diseases. We explored the use of a recombinant measles virus (MV) hTERT vector to boost DNA priming as recombinant live attenuated measles virus has an impressive safety and efficacy record. Here, we show that a MV-TERT vector can rapidly and strongly boost DNA hTERT priming in MV susceptible IFNAR/CD46 mouse models. The cellular immune responses were Th1 polarized. No humoral responses were elicited. The 4 kb hTERT transgene did not impact MV replication or induction of cell-mediated responses. These findings validate the MV-TERT vector to boost cell-mediated responses following DNA priming in humans.


Assuntos
Vacinas Anticâncer/imunologia , Epitopos de Linfócito T/imunologia , Vetores Genéticos , Imunidade Celular , Vírus do Sarampo , Linfócitos T/imunologia , Telomerase/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Vacinas Anticâncer/genética , Linhagem Celular , Chlorocebus aethiops , Citocinas/metabolismo , Citotoxicidade Imunológica , Vetores Genéticos/genética , Humanos , Imunização , Imunização Secundária , Vírus do Sarampo/genética , Camundongos , Camundongos Transgênicos , Telomerase/genética , Vacinas de DNA , Células Vero
4.
Nucleic Acids Res ; 45(6): 3231-3241, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-28100701

RESUMO

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.


Assuntos
Citidina Desaminase/biossíntese , Quebras de DNA de Cadeia Dupla , DNA Mitocondrial/metabolismo , Linhagem Celular Tumoral , Cromossomos Humanos , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Citosol/metabolismo , Proteína DEAD-box 58 , DNA Mitocondrial/química , Humanos , Interferon-alfa/biossíntese , Interferon beta/biossíntese , Interferon beta/fisiologia , Proteínas/genética , Proteínas/metabolismo , RNA Polimerase III/metabolismo , Receptores Imunológicos , Transcrição Gênica , Regulação para Cima , Uracila/metabolismo
5.
Nucleic Acids Res ; 43(19): 9340-9, 2015 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-26384561

RESUMO

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.


Assuntos
Citidina Desaminase/genética , Quebras de DNA de Cadeia Dupla , Animais , Linhagem Celular , Citidina Desaminase/química , Citidina Desaminase/metabolismo , Células HeLa , Humanos , Macaca mulatta , Antígenos de Histocompatibilidade Menor , Mutação , Fenótipo , Estrutura Terciária de Proteína , Proteínas/química , Proteínas/genética , Codorniz , Edição de RNA
6.
Retrovirology ; 13(1): 84, 2016 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-27998270

RESUMO

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.


Assuntos
DNA Circular/análise , DNA Viral/análise , Vírus da Hepatite B/genética , Vírus da Hepatite B/isolamento & purificação , Hepatite B/virologia , Reação em Cadeia da Polimerase em Tempo Real , DNA Circular/sangue , DNA Viral/sangue , DNA Viral/genética , Vírus da Hepatite B/fisiologia , Hepatite B Crônica/virologia , Humanos , Vírion/genética , Replicação Viral
7.
Mol Biol Evol ; 31(2): 330-40, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24162735

RESUMO

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.


Assuntos
Citidina Desaminase/metabolismo , Citidina/análogos & derivados , DNA/genética , Mamíferos/metabolismo , Sequência de Aminoácidos , Animais , Citidina/metabolismo , Citidina Desaminase/química , Citidina Desaminase/genética , Quebras de DNA de Cadeia Dupla , Desaminação , Cães , Instabilidade Genômica , Células HEK293 , Células HeLa , Humanos , Células Madin Darby de Rim Canino , Mamíferos/genética , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Filogenia , Estrutura Secundária de Proteína , Proteínas/química , Proteínas/genética , Proteínas/metabolismo , Alinhamento de Sequência
9.
Proc Natl Acad Sci U S A ; 108(12): 4858-63, 2011 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-21368204

RESUMO

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.


Assuntos
Citosina Desaminase/metabolismo , DNA Mitocondrial/metabolismo , Loci Gênicos , Genoma Humano , Mutação , Desaminases APOBEC , Citidina Desaminase , Citosina Desaminase/genética , DNA Mitocondrial/genética , Feminino , Células HeLa , Hepadnaviridae/genética , Hepadnaviridae/metabolismo , Humanos , Masculino , Neoplasias/enzimologia , Neoplasias/genética , Retroviridae/genética , Retroviridae/metabolismo , Uracila-DNA Glicosidase/deficiência , Uracila-DNA Glicosidase/genética , Uracila-DNA Glicosidase/metabolismo
10.
J Biol Chem ; 287(46): 39182-92, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-22977230

RESUMO

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.


Assuntos
Núcleo Celular/metabolismo , Citidina Desaminase/metabolismo , Citidina/química , DNA/metabolismo , Regulação da Expressão Gênica , Proteínas/metabolismo , Ciclo Celular , Sobrevivência Celular , Citidina Desaminase/fisiologia , Reparo do DNA , Desaminação , Genoma , Células HEK293 , Células HeLa , Humanos , Microscopia Confocal/métodos , Mutagênese , Mutação , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas/fisiologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Técnicas do Sistema de Duplo-Híbrido
11.
J Virol ; 86(16): 8592-601, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22674981

RESUMO

Herpes simplex virus 1 (HSV-1) is a human pathogen that leads to recurrent facial-oral lesions. Its 152-kb genome is organized in two covalently linked segments, each composed of a unique sequence flanked by inverted repeats. Replication of the HSV-1 genome produces concatemeric molecules in which homologous recombination events occur between the inverted repeats. This mechanism leads to four genome isomers (termed P, IS, IL, and ILS) that differ in the relative orientations of their unique fragments. Molecular combing analysis was performed on DNA extracted from viral particles and BSR, Vero, COS-7, and Neuro-2a cells infected with either strain SC16 or KOS of HSV-1, as well as from tissues of experimentally infected mice. Using fluorescence hybridization, isomers were repeatedly detected and distinguished and were accompanied by a large proportion of noncanonical forms (40%). In both cell and viral-particle extracts, the distributions of the four isomers were statistically equivalent, except for strain KOS grown in Vero and Neuro-2a cells, in which P and IS isomers were significantly overrepresented. In infected cell extracts, concatemeric molecules as long as 10 genome equivalents were detected, among which, strikingly, the isomer distributions were equivalent, suggesting that any such imbalance may occur during encapsidation. In vivo, for strain KOS-infected trigeminal ganglia, an unbalanced distribution distinct from the one in vitro was observed, along with a considerable proportion of noncanonical assortment.


Assuntos
Genoma Viral , Herpesvirus Humano 1/genética , Polimorfismo Genético , Animais , Linhagem Celular , DNA Viral/genética , DNA Viral/isolamento & purificação , Modelos Animais de Doenças , Herpes Simples/virologia , Herpesvirus Humano 1/crescimento & desenvolvimento , Herpesvirus Humano 1/isolamento & purificação , Humanos , Camundongos , Hibridização de Ácido Nucleico
12.
J Gen Virol ; 93(Pt 12): 2646-2651, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22993189

RESUMO

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.


Assuntos
Adenosina Desaminase/metabolismo , Vírus Linfotrópico T Tipo 2 Humano/genética , Vírus Linfotrópico T Tipo 2 Humano/metabolismo , Edição de RNA/fisiologia , RNA Viral/genética , RNA Viral/metabolismo , Vírus Linfotrópico T Tipo 3 de Símios/genética , Vírus Linfotrópico T Tipo 3 de Símios/metabolismo , Adenosina/química , Adenosina Desaminase/genética , Animais , Sequência de Bases , Genoma Viral , Células HEK293 , Humanos , Dados de Sequência Molecular , Mutação , Reação em Cadeia da Polimerase/métodos , RNA Viral/química , Proteínas de Ligação a RNA , Homologia de Sequência do Ácido Nucleico
13.
J Virol ; 85(23): 12227-40, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21917974

RESUMO

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.


Assuntos
Antivirais/farmacologia , Vírus da Febre Aftosa/genética , Genes Letais , Genoma Viral , Mutagênese , Mutação/genética , Ribavirina/farmacologia , Animais , Células Cultivadas , Cricetinae , Replicação do DNA , Farmacorresistência Viral , Vírus da Febre Aftosa/efeitos dos fármacos , Vírus da Febre Aftosa/crescimento & desenvolvimento , Rim/citologia , Taxa de Mutação , RNA Mensageiro/genética , RNA Viral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Replicação Viral
14.
J Virol ; 85(15): 7594-602, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21632763

RESUMO

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.


Assuntos
Citosina Desaminase/metabolismo , Genoma Viral , Herpesvirus Humano 1/genética , Herpesvirus Humano 4/genética , Desaminases APOBEC , Animais , Sequência de Bases , Chlorocebus aethiops , Citidina Desaminase , Citosina Desaminase/genética , DNA/genética , Células HeLa , Herpesvirus Humano 1/fisiologia , Herpesvirus Humano 4/fisiologia , Humanos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Homologia de Sequência do Ácido Nucleico , Células Vero , Replicação Viral
15.
J Virol ; 85(5): 2458-62, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21159878

RESUMO

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.


Assuntos
Adenosina Desaminase/metabolismo , Genoma Viral , Vírus da Influenza A/genética , Influenza Humana/enzimologia , Vírus do Sarampo/genética , Sarampo/enzimologia , Mutação , Adenosina Desaminase/genética , Animais , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , Humanos , Vacinas contra Influenza/genética , Influenza Humana/virologia , Sarampo/virologia , Vacina contra Sarampo/genética , Dados de Sequência Molecular , Proteínas de Ligação a RNA , Estações do Ano , Vacinas Atenuadas/genética , Células Vero
16.
PLoS Pathog ; 6(5): e1000928, 2010 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-20523896

RESUMO

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.


Assuntos
Citidina Desaminase/genética , DNA Viral/genética , Vírus da Hepatite B/genética , Hepatite B/imunologia , Hepatite B/virologia , Desaminase APOBEC-3G , Idoso , Citidina Desaminase/metabolismo , DNA de Cadeia Simples/genética , Evolução Molecular , Feminino , Regulação Viral da Expressão Gênica/imunologia , Genoma Viral , Antígenos do Núcleo do Vírus da Hepatite B/genética , Vacinas contra Hepatite B/genética , Vacinas contra Hepatite B/imunologia , Vírus da Hepatite B/imunologia , Humanos , Cirrose Hepática/virologia , Masculino , Pessoa de Meia-Idade , Mutação , Regulação para Cima/fisiologia
17.
Blood ; 115(15): 3025-32, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20130242

RESUMO

The success of active immunotherapy is based on the vaccine's ability to overcome immune tolerance through recalibrating the immune system so that it is able to recognize tumor antigens as foreign rather than self. In this study, we used a lentiviral vector system to target human telomerase reverse transcriptase (lv-hTERT), a widely expressed tumor antigen. Immunization of HLA-A*0201 transgenic HHD mice with recombinant lv-hTERT vector induces potent and diversified cytotoxic T lymphocyte responses that recognize in vitro murine tumor cells, which overexpress telomerase. Compared with peptide-based vaccinations, the lv-hTERT vector triggers better and more sustained CD8(+) T-cell response against self/TERT epitope in vivo. The study found that the additional use of a heterologous boosted vaccination drastically improves self/TERT-specific CD8 responses in lv-hTERT primed mice. Both primary and long-lasting self/TERT-specific CD8(+) T-cell responses induced with Iv-hTERT vector required the presence of CD4 T cells in vivo. This lv-hTERT-based active immunotherapy efficiently inhibits the growth of telomerase expressing tumors (B16/HLA-A2.1 murine melanoma) in HHD mice. These data show that targeting hTERT with lentivector is highly effective in stimulating a broad range of CD8 T-cell immunity that can be exploited for cancer immunotherapy.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Terapia Genética , Vetores Genéticos/genética , Imunidade Celular/imunologia , Lentivirus/genética , Neoplasias/terapia , Telomerase/uso terapêutico , Animais , Linfócitos T CD8-Positivos/citologia , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Proliferação de Células , Apresentação Cruzada/imunologia , Humanos , Imunização , Depleção Linfocítica , Camundongos , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/prevenção & controle , Peptídeos/imunologia , Recombinação Genética , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/imunologia , Telomerase/genética
19.
Science ; 375(6586): 1235, 2022 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-35298253

RESUMO

Discoverer of the human immunodeficiency virus.

20.
J Virol ; 84(14): 7278-87, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20463074

RESUMO

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.


Assuntos
Citidina Desaminase/metabolismo , Genoma Viral , Infecções por HTLV-I/virologia , Vírus Linfotrópico T Tipo 1 Humano/genética , Leucemia-Linfoma de Células T do Adulto/virologia , Mutação , Provírus/genética , Desaminase APOBEC-3G , Sequência de Bases , Linhagem Celular , Citidina Desaminase/genética , Genes Reporter , Variação Genética , Vetores Genéticos , Vírus Linfotrópico T Tipo 1 Humano/imunologia , Vírus Linfotrópico T Tipo 1 Humano/patogenicidade , Humanos , Dados de Sequência Molecular , Mutagênese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA