Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 86(9): 5393-8, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22379085

RESUMO

The liver-enriched transcriptional activator protein (LAP) isoform of CCAAT/enhancer binding protein ß (C/EBPß) is shown to be a major activator of differentiation-dependent human papillomavirus (HPV) late gene expression, while the liver-enriched inhibitory protein (LIP) isoform negatively regulates late expression. In undifferentiated cells, LIPs act as dominant-negative repressors of late expression, and upon differentiation, LIP levels are significantly reduced, allowing LAP-mediated activation of the late promoter. Importantly, knockdown of C/EBPß isoforms blocks activation of late gene expression from complete viral genomes upon differentiation.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Diferenciação Celular/genética , Regulação Viral da Expressão Gênica , Papillomavirus Humano 31/genética , Papillomavirus Humano 31/metabolismo , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Inativação Gênica , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Mutagênese Sítio-Dirigida , Regiões Promotoras Genéticas , Isoformas de Proteínas/metabolismo , Elementos de Resposta , Transcrição Gênica
2.
J Virol ; 84(19): 10209-19, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20686027

RESUMO

Tandem stop mutations K26X and H27X in human immunodeficiency virus type 1 (HIV-1) vif compromise virus replication in human T-cell lines that stably express APOBEC3F (A3F) or APOBEC3G (A3G). We previously reported that partial resistance to A3G could develop in these Vif-deficient viruses through a nucleotide A200-to-T/C transversion and a vpr null mutation, but these isolates were still susceptible to restriction by A3F. Here, long-term selection experiments were done to determine how these A3G-selected isolates might evolve to spread in the presence of A3F. We found that A3F, like A3G, is capable of potent, long-term restriction that eventually selects for heritable resistance. In all 7 instances, the selected isolates had restored Vif function to cope with A3F activity. In two isolates, Vif Q26-Q27 and Y26-Q27, the resistance phenotype recapitulated in molecular clones, but when the selected vif alleles were analyzed in the context of an otherwise wild-type viral background, a different outcome emerged. Although HIV-1 clones with Vif Q26-Q27 or Y26-Q27 were fully capable of overcoming A3F, they were now susceptible to restriction by A3G. Concordant with prior studies, a lysine at position 26 proved essential for A3G neutralization. These data combine to indicate that A3F and A3G exert at least partly distinct selective pressures and that Vif function may be essential for the virus to replicate in the presence of A3F.


Assuntos
Citosina Desaminase/fisiologia , HIV-1/genética , HIV-1/fisiologia , Produtos do Gene vif do Vírus da Imunodeficiência Humana/genética , Produtos do Gene vif do Vírus da Imunodeficiência Humana/fisiologia , Desaminase APOBEC-3G , Alelos , Sequência de Bases , Linhagem Celular , Citidina Desaminase/fisiologia , Primers do DNA/genética , DNA Viral/genética , Genes vif , HIV-1/patogenicidade , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Mutagênese Sítio-Dirigida , Mutação , Seleção Genética , Linfócitos T/fisiologia , Linfócitos T/virologia , Replicação Viral/genética , Replicação Viral/fisiologia
4.
J Virol ; 83(11): 5956-60, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19297501

RESUMO

APOBEC3G restricts Vif-deficient human immunodeficiency virus type 1 (HIV-1) by deaminating viral cDNA cytosines to uracils. This promutagenic activity is counteracted by HIV-1 Vif, which is a natural APOBEC3G antagonist. However, we previously reported that Vif-deficient HIV-1 could evolve resistance to APOBEC3G by a novel mechanism requiring an A200-to-C/T transition mutation and Vpr inactivation. A pyrimidine at nucleotide 200 in the untranslated leader region contributed to resistance by increasing virus particle production, which resulted in fewer APOBEC3G molecules per particle. Here we show that the A200-to-C/T mutation functions posttranscriptionally by inactivating an upstream start codon, which in turn enables optimal viral mRNA translation from canonical start codons.


Assuntos
Citidina Desaminase/metabolismo , Produtos do Gene vif/deficiência , HIV-1/metabolismo , Biossíntese de Proteínas/genética , Desaminase APOBEC-3G , Sequência de Bases , Citidina Desaminase/genética , Produtos do Gene vif/genética , Genótipo , HIV-1/genética , Humanos , Cinética , Dados de Sequência Molecular
5.
Curr Biol ; 18(11): 819-24, 2008 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-18501607

RESUMO

The human APOBEC3G protein restricts the replication of Vif-deficient HIV-1 by deaminating nascent viral cDNA cytosines to uracils, leading to viral genomic strand G-to-A hypermutations. However, the HIV-1 Vif protein triggers APOBEC3G degradation, which helps to explain why this innate defense does not protect patients. The APOBEC3G-Vif interaction is a promising therapeutic target, but the benefit of the enabling of HIV-1 restriction in patients is unlikely to be known until Vif antagonists are developed. As a necessary prelude to such studies, cell-based HIV-1 evolution experiments were done to find out whether APOBEC3G can provide a long-term block to Vif-deficient virus replication and, if so, whether HIV-1 variants that resist restriction would emerge. APOBEC3G-expressing T cells were infected with Vif-deficient HIV-1. Virus infectivity was suppressed in 45/48 cultures for more than five weeks, but replication was eventually detected in three cultures. Virus-growth characteristics and sequencing demonstrated that these isolates were still Vif-deficient and that in fact, these viruses had acquired a promoter mutation and a Vpr null mutation. Resistance occurred by a novel tolerance mechanism in which the resistant viruses packaged less APOBEC3G and accumulated fewer hypermutations. These data support the development of antiretrovirals that antagonize Vif and thereby enable endogenous APOBEC3G to suppress HIV-1 replication.


Assuntos
Evolução Biológica , Citidina Desaminase/metabolismo , HIV-1/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Produtos do Gene vif do Vírus da Imunodeficiência Humana/metabolismo , Desaminase APOBEC-3G , Humanos , Replicação Viral/fisiologia
6.
J Virol ; 82(6): 2652-60, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18184715

RESUMO

Human APOBEC3G and several other APOBEC3 proteins have been shown to inhibit the replication of a variety of retrotransposons and retroviruses. All of these enzymes can deaminate cytosines within single-strand DNA, but the overall importance of this conserved activity in retroelement restriction has been questioned by reports of deaminase-independent mechanisms. Here, three distinct retroelements, a yeast retrotransposon, Ty1, a murine endogenous retrovirus, MusD, and a lentivirus, human immunodeficiency virus type 1 (HIV-1), were used to evaluate the relative contributions of deaminase-dependent and -independent mechanisms. Although human APOBEC3G can restrict the replication of all three of these retroelements, APOBEC3G lacking the catalytic glutamate (E259Q) was clearly defective. This phenotype was particularly clear in experiments with low levels of APOBEC3G expression. In contrast, purposeful overexpression of APOBEC3G-E259Q was able to cause modest to severe reductions in the replication of Ty1, MusD, and HIV-1(DeltaVif). The importance of these observations was highlighted by data showing that CEM-SS T-cell lines expressing near-physiologic levels of APOBEC3G-E259Q failed to inhibit the replication of HIV-1(DeltaVif), whereas similar levels of wild-type APOBEC3G fully suppressed virus infectivity. Despite the requirement for DNA deamination, uracil DNA glycosylase did not modulate APOBEC3G-dependent restriction of Ty1 or HIV-1(DeltaVif), further supporting prior studies indicating that the major uracil excision repair system of cells is not involved. In conclusion, the absolute requirement for the catalytic glutamate of APOBEC3G in Ty1, MusD, and HIV-1 restriction strongly indicates that DNA cytosine deamination is an essential part of the mechanism.


Assuntos
Citidina Desaminase/metabolismo , HIV-1/fisiologia , Desaminase APOBEC-3G , Sequência de Bases , Linhagem Celular , Citidina Desaminase/genética , Primers do DNA , HIV-1/patogenicidade , Humanos , Mutagênese Sítio-Dirigida , Zinco/metabolismo
7.
AIDS Rev ; 8(3): 148-57, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17078485

RESUMO

Over 40 million people worldwide currently have HIV/AIDS. Many antiretroviral drugs have proven effective, but drug-resistant HIV variants frequently emerge to thwart treatment efforts. Reverse transcription errors undoubtedly contribute to drug resistance, but additional significant sources of viral genetic variation are debatable. The human APOBEC3F and APOBEC3G proteins can potently inhibit retrovirus infection by a mechanism that involves retroviral cDNA cytosine deamination. Here we review the current knowledge on the mechanism of APOBEC3-dependent retrovirus restriction and discuss whether this innate host-defense system actively contributes to HIV genetic variation.


Assuntos
Citosina Desaminase/metabolismo , Farmacorresistência Viral Múltipla/genética , Infecções por HIV/tratamento farmacológico , HIV/efeitos dos fármacos , Imunidade Inata/imunologia , Infecções por Retroviridae/prevenção & controle , Desaminases APOBEC , Citidina Desaminase , Farmacorresistência Viral Múltipla/efeitos dos fármacos , Variação Genética/imunologia , Infecções por HIV/genética , Humanos
8.
Nucleic Acids Res ; 34(19): 5683-94, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17038330

RESUMO

The APOBEC3 proteins are unique to mammals. Many inhibit retrovirus infection through a cDNA cytosine deamination mechanism. HIV-1 neutralizes this host defense through Vif, which triggers APOBEC3 ubiquitination and degradation. Here, we report an APOBEC3F-like, double deaminase domain protein from three artiodactyls: cattle, pigs and sheep. Like their human counterparts, APOBEC3F and APOBEC3G, the artiodactyl APOBEC3F proteins are DNA cytosine deaminases that locate predominantly to the cytosol and can inhibit the replication of HIV-1 and MLV. Retrovirus restriction is attributable to deaminase-dependent and -independent mechanisms, as deaminase-defective mutants retain significant anti-retroviral activity. However, unlike human APOBEC3F and APOBEC3G, the artiodactyl APOBEC3F proteins have an active N-terminal DNA cytosine deaminase domain, which elicits a broader dinucleotide deamination preference, and they are resistant to HIV-1 Vif. These data indicate that DNA cytosine deamination; sub-cellular localization and retrovirus restriction activities are conserved in mammals, whereas active site location, local mutational preferences and Vif susceptibility are not. Together, these studies indicate that some properties of the mammal-specific, APOBEC3-dependent retroelement restriction system are necessary and conserved, but others are simultaneously modular and highly adaptable.


Assuntos
Artiodáctilos/genética , Citosina Desaminase/química , Evolução Molecular , Retroviridae/genética , Sequência de Aminoácidos , Animais , Catálise , Bovinos , Citoplasma/enzimologia , Citosina Desaminase/genética , Citosina Desaminase/metabolismo , Desaminação , Produtos do Gene vif/metabolismo , Células HeLa , Humanos , Dados de Sequência Molecular , Mutação , Filogenia , Estrutura Terciária de Proteína , Alinhamento de Sequência , Ovinos/genética , Suínos/genética , Zinco/química
9.
J Biol Chem ; 280(12): 10920-4, 2005 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-15647250

RESUMO

The human proteins APOBEC3F and APOBEC3G restrict retroviral infection by deaminating cytosine residues in the first cDNA strand of a replicating virus. These proteins have two putative deaminase domains, and it is unclear whether one or both catalyze deamination, unlike their homologs, AID and APOBEC1, which are well characterized single domain deaminases. Here, we show that only the C-terminal cytosine deaminase domain of APOBEC3F and -3G governs retroviral hypermutation. A chimeric protein with the N-terminal cytosine deaminase domain from APOBEC3G and the C-terminal cytosine deaminase domain from APOBEC3F elicited a dinucleotide hypermutation preference nearly indistinguishable from that of APOBEC3F. This 5'-TC-->TT mutational specificity was confirmed in a heterologous Escherichia coli-based mutation assay, in which the 5'-CC-->CT dinucleotide hypermutation preference of APOBEC3G also mapped to the C-terminal deaminase domain. An N-terminal APOBEC3G deletion mutant displayed a preference indistinguishable from that of the full-length protein, and replacing the C-terminal deaminase domain of APOBEC3F with AID resulted in an AID-like mutational signature. Together, these data indicate that only the C-terminal domain of APOBEC3F and -3G dictates the retroviral minus strand 5'-TC and 5'-CC dinucleotide hypermutation preferences, respectively, leaving the N-terminal domain to perform other aspects of retroviral restriction.


Assuntos
Citosina Desaminase/química , Mutação , Proteínas/química , Retroviridae/genética , Desaminase APOBEC-3G , Sequência de Bases , Citidina Desaminase , Citosina Desaminase/fisiologia , Dados de Sequência Molecular , Nucleosídeo Desaminases , Proteínas/fisiologia , Proteínas Repressoras
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...