Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 162
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 79(15): 9579-87, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16014920

RESUMO

The human cytidine deaminase Apobec3F (h-A3F), a protein related to the previously recognized antiviral factor Apobec3G (h-A3G), has antiviral activity against human immunodeficiency virus type 1 (HIV-1) that is suppressed by the viral protein Vif. The mechanism of HIV-1 Vif-mediated suppression of h-A3F is not fully understood. Here, we demonstrate that while h-A3F, like h-A3G, was able to suppress primate lentiviruses other than HIV-1 (simian immunodeficiency virus from African green monkeys [SIVagm] and Rhesus macaques [SIVmac]), the interaction between Vif proteins and h-A3F appeared to differ from that with h-A3G. H-A3F showed no change in its species specificity against HIV-1 or SIVagm Vif when a negatively charged amino acid was replaced with a lysine at position 128, a residue critical for h-A3G recognition by HIV-1 Vif. However, HIV-1 Vif, but not SIVagm Vif, was able to bind h-A3F and induce its polyubiquitination and degradation through the Cul5-containing E3 ubiquitin ligase. Interference with Cul5-E3 ligase function by depletion of Cul5, through RNA interference or overexpression of Cul5 mutants, blocked the ability of HIV-1 Vif to suppress h-A3F. A BC-box mutant of HIV-1 Vif that failed to recruit Cul5-E3 ligase but was still able to interact with h-A3F failed to suppress h-A3F. Interestingly, interference with Cul5-E3 ligase function or overexpression of h-A3F or h-A3G also increased the stability of HIV-1 Vif, suggesting that like the substrate molecules h-A3F and h-A3G, the substrate receptor protein Vif is itself also regulated by Cul5-E3 ligase. Our results indicate that Cul5-E3 ligase appears to be a common pathway hijacked by HIV-1 Vif to defeat both h-A3F and h-A3G. Developing inhibitors to disrupt the interaction between Vif and Cul5-E3 ligase could be therapeutically useful, allowing multiple host antiviral factors to suppress HIV-1.


Assuntos
Antivirais/fisiologia , Proteínas Culina/metabolismo , Citosina Desaminase/fisiologia , Produtos do Gene vif/metabolismo , HIV-1/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Antivirais/metabolismo , Linhagem Celular , Proteínas Culina/genética , Citosina Desaminase/metabolismo , Deleção de Genes , Humanos , Lentivirus de Primatas/fisiologia , Complexos Ubiquitina-Proteína Ligase/metabolismo , Replicação Viral , Produtos do Gene vif do Vírus da Imunodeficiência Humana
2.
Hepatology ; 42(2): 301-9, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16025511

RESUMO

APOBEC3G is a cellular cytidine deaminase displaying broad antiretroviral activity. Recently, it was shown that APOBEC3G can also suppress hepatitis B virus (HBV) production in human hepatoma cells. In the present study, we characterized the mechanisms of APOBEC-mediated antiviral activity against HBV and related hepadnaviruses. We show that human APOBEC3G blocks HBV production in mammalian and nonmammalian cells and is active against duck HBV as well. Early steps of viral morphogenesis, including RNA and protein synthesis, binding of pregenomic RNA to core protein, and self-assembly of viral core protein, were unaffected. However, APOBEC3G rendered HBV core protein-associated full-length pregenomic RNA nuclease-sensitive. Ongoing reverse-transcription in capsids that had escaped the block in morphogenesis was not significantly inhibited. The antiviral effect was not modulated by abrogating or enhancing expression of the accessory HBV X protein, suggesting that HBV X protein does not represent a functional homologue to the HIV vif protein. Furthermore, human APOBEC3F but not rat APOBEC1 inhibited HBV DNA production. Viral RNA and low-level DNA produced in the presence of APOBEC3F or rat APOBEC1 occasionally displayed mutations, but the majority of clones were wild-type. In conclusion, APOBEC3G and APOBEC3F but not rat APOBEC1 can downregulate the production of replication-competent hepadnaviral nucleocapsids. In contrast to HIV and other retroviruses, however, APOBEC3G/3F-mediated editing of nucleic acids does not seem to represent an effective innate defense mechanism for hepadnaviruses.


Assuntos
Antivirais/fisiologia , Citosina Desaminase/fisiologia , Vírus da Hepatite B do Pato/fisiologia , Vírus da Hepatite B/fisiologia , Proteínas/fisiologia , Desaminase APOBEC-1 , Desaminase APOBEC-3G , Animais , Galinhas , Citidina Desaminase/fisiologia , DNA Viral/biossíntese , Vírus da Hepatite B/genética , Nucleosídeo Desaminases , DNA Polimerase Dirigida por RNA/metabolismo , Ratos , Proteínas Repressoras , Proteínas do Core Viral/biossíntese , Montagem de Vírus
3.
J Virol ; 79(14): 8969-78, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15994791

RESUMO

The tripartite motif 5alpha protein (TRIM5alpha) is one of several factors expressed by mammalian cells that inhibit retrovirus replication. Human TRIM5alpha (huTRIM5alpha) inhibits infection by N-tropic murine leukemia virus (N-MLV) but is inactive against human immunodeficiency virus type 1 (HIV-1). However, we show that replacement of a small segment in the carboxy-terminal B30.2/SPRY domain of huTRIM5alpha with its rhesus macaque counterpart (rhTRIM5alpha) endows it with the ability to potently inhibit HIV-1 infection. The B30.2/SPRY domain and an additional domain in huTRIM5alpha, comprising the amino-terminal RING and B-box components of the TRIM motif, are required for N-MLV restriction activity, while the intervening coiled-coil domain is necessary and sufficient for huTRIM5alpha multimerization. Truncated huTRIM5alpha proteins that lack either or both the N-terminal RING/B-Box or the C-terminal B30.2/SPRY domain form heteromultimers with full-length huTRIM5alpha and are dominant inhibitors of its N-MLV restricting activity, suggesting that homomultimerization of intact huTRIM5alpha monomers is necessary for N-MLV restriction. However, localization in large cytoplasmic bodies is not required for inhibition of N-MLV by huTRIM5alpha or for inhibition of HIV-1 by chimeric or rhTRIM5alpha.


Assuntos
Antivirais/fisiologia , Proteínas de Transporte/química , Proteínas de Transporte/fisiologia , Infecções por Retroviridae/prevenção & controle , Sequência de Aminoácidos , Fatores de Restrição Antivirais , HIV-1/fisiologia , Células HeLa , Humanos , Vírus da Leucemia Murina/fisiologia , Dados de Sequência Molecular , Relação Estrutura-Atividade , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases
4.
Biochem Biophys Res Commun ; 331(4): 1358-64, 2005 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-15883025

RESUMO

Human respiratory syncytial virus (hRSV) membrane fusion is promoted by the formation of a trimer-of-hairpins structure that brings the amino- and carboxyl-terminal regions of fusion (F) protein into close proximity. Two heptad-repeat (HR1 and HR2) regions in F protein play an important role in this process. Our previous study demonstrated that peptides derived from HR1 and HR2 regions of F protein were potent inhibitors of hRSV entry. Here we showed that HR1 peptide and its analog denoted 5-Helix which contained a central coiled-coil formed by three HR1s could induce highly potent antibody response in the immunized rabbits. Both antibodies could recognize F1 domain of the F protein and inhibited hRSV entry with the neutralizing antibody titers of 1:61 and 1:115, respectively. These suggested that 5-Helix could induce potent neutralizing antibody response and the central coiled-coil might be a highly conserved neutralization site for hRSV F protein.


Assuntos
Anticorpos/imunologia , Antivirais/fisiologia , Fragmentos de Peptídeos/imunologia , Vírus Sinciciais Respiratórios/metabolismo , Proteínas Virais de Fusão/química , Sequência de Aminoácidos , Linhagem Celular , Eletroforese em Gel de Poliacrilamida , Soros Imunes , Dados de Sequência Molecular , Testes de Neutralização
6.
Science ; 308(5721): 557-60, 2005 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-15845854

RESUMO

In eukaryotes, 21- to 24-nucleotide-long RNAs engage in sequence-specific interactions that inhibit gene expression by RNA silencing. This process has regulatory roles involving microRNAs and, in plants and insects, it also forms the basis of a defense mechanism directed by small interfering RNAs that derive from replicative or integrated viral genomes. We show that a cellular microRNA effectively restricts the accumulation of the retrovirus primate foamy virus type 1 (PFV-1) in human cells. PFV-1 also encodes a protein, Tas, that suppresses microRNA-directed functions in mammalian cells and displays cross-kingdom antisilencing activities. Therefore, through fortuitous recognition of foreign nucleic acids, cellular microRNAs have direct antiviral effects in addition to their regulatory functions.


Assuntos
Antivirais/fisiologia , MicroRNAs/fisiologia , Interferência de RNA , Spumavirus/genética , Spumavirus/fisiologia , Animais , Arabidopsis/genética , Linhagem Celular , Cricetinae , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Genes Reporter , Proteínas de Fluorescência Verde/genética , Células HeLa , Humanos , Oligonucleotídeos Antissenso , Plantas Geneticamente Modificadas , Biossíntese de Proteínas , RNA Viral , Proteínas dos Retroviridae/genética , Proteínas dos Retroviridae/metabolismo , Transativadores/genética , Transativadores/metabolismo , Transfecção , Replicação Viral
7.
Eur J Med Res ; 10(3): 117-20, 2005 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-15851378

RESUMO

BACKGROUND: Severe acute respiratory syndrome (SARS) is a novel infectious disease which is characterized by an overaggressive immune response. Chemokines are important inflammatory mediators and regulate disease due to viral infection. In previous study, we found that SARS-CoV has the ability to replicate in mononuclear cells. In present work, we sought to characterize the replication of SARS-CoV at the presence of RANTES in THP-1 cells. METHODS: To determine whether RANTES play an role in the process of SARS, THP-1 cells were incubated with heat-inactivated SARS-CoV and ELISA was used to test RANTES levels in the supernatants; Then the effect of dexamethasone on the induced secretion was evaluated. Real-time PCR was used to investigate the effort of RANTES on the replication of SARS-CoV in vitro. Macrophages, induced by THP-1 cells, were used as cell model. FINDINGS: Inactive SARS-CoV could induce THP-1 cells secret RANTES and this increase effect could not be suppressed by DXM. RANTES itself could inhibit the replication of SARS-CoV in THP-1 cells when it was added into the culture before or at the same time with the virus; No inhibition effect was shown when RANTES were added into the culture after SARS-CoV infected the cells.


Assuntos
Antivirais/fisiologia , Quimiocina CCL5/fisiologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Replicação Viral/fisiologia , Animais , Células CHO , Linhagem Celular Tumoral , Chlorocebus aethiops , Cricetinae , Humanos , Macrófagos/virologia , Células Vero
8.
J Immunol ; 174(4): 1932-7, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15699120

RESUMO

Toll-like receptors (TLRs) mediate host cell activation by various microbial components. TLR2, TLR3, TLR4, TLR7, TLR8, and TLR9 are the receptors that have been associated with virus-induced immune response. We have previously reported that all these TLRs, except TLR9, are expressed at mRNA levels in human monocyte-derived macrophages. Here we have studied TLR2, TLR3, TLR4, and TLR7/8 ligand-induced IFN-alpha, IFN-beta, IL-28, and IL-29 expression in human macrophages. IFN-alpha pretreatment of macrophages was required for efficient TLR3 and TLR4 agonist-induced activation of IFN-alpha, IFN-beta, IL-28, and IL-29 genes. TLR7/8 agonist weakly activated IFN-alpha, IFN-beta, IL-28, and IL-29 genes, whereas TLR2 agonist was not able to activate these genes. IFN-alpha enhanced TLR responsiveness in macrophages by up-regulating the expression of TLR3, TLR4, and TLR7. IFN-alpha also enhanced the expression of TLR signaling molecules MyD88, TIR domain-containing adaptor inducing IFN-beta, IkappaB kinase-epsilon, receptor interacting protein 1, and IFN regulatory factor 7. Furthermore, the activation of transcription factor IFN regulatory factor 3 by TLR3 and TLR4 agonists was dependent on IFN-alpha pretreatment. In conclusion, our results suggest that IFN-alpha sensitizes cells to microbial recognition by up-regulating the expression of several TLRs as well as adapter molecules and kinases involved in TLR signaling.


Assuntos
Regulação da Expressão Gênica/imunologia , Interferon-alfa/fisiologia , Interferon beta/genética , Interleucinas/genética , Glicoproteínas de Membrana/fisiologia , Receptores de Superfície Celular/fisiologia , Antivirais/fisiologia , Linhagem Celular , Sistema Livre de Células/imunologia , Células Cultivadas , Citocinas , Proteínas de Ligação a DNA/metabolismo , Humanos , Vírus da Influenza A/imunologia , Fator Regulador 1 de Interferon , Interferon-alfa/biossíntese , Interferon-alfa/genética , Interferon beta/biossíntese , Interferon gama/biossíntese , Interferons , Interleucinas/biossíntese , Ligantes , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/virologia , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/biossíntese , NF-kappa B/metabolismo , Fosfoproteínas/metabolismo , Receptores de Superfície Celular/agonistas , Receptores de Superfície Celular/biossíntese , Vírus Sendai/imunologia , Transdução de Sinais/imunologia , Receptor 2 Toll-Like , Receptor 3 Toll-Like , Receptor 4 Toll-Like , Receptor 7 Toll-Like , Receptor 8 Toll-Like , Receptor Toll-Like 9 , Receptores Toll-Like
9.
J Immunol ; 174(3): 1574-9, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15661918

RESUMO

CD8(+) cells from HIV-infected individuals showing the CD8(+) cell noncytotoxic antiviral response unexpectedly revealed mRNA for VCAM-1, a cell surface molecule found on endothelial cells. Uninfected subjects had undetectable levels of VCAM-1 mRNA in their CD8(+) cells. Flow cytometry analysis showed that up to 12% of the CD8(+) cells from HIV-positive individuals expressed VCAM-1 compared with 0.8% of the CD8(+) cells of HIV-negative individuals. Enrichment of the CD8(+)VCAM-1(+) cell population and subsequent coculture with CD4(+) cells acutely infected with HIV-1 showed that the VCAM-1(+)CD8(+) cells were able to suppress viral replication with 50% less input cells than the unseparated CD8(+) cell population. This study demonstrates, for the first time, the expression of VCAM-1 on CD8(+) cells. Moreover, the CD8(+)VCAM-1(+) cells show enhanced CD8(+) cell noncytotoxic antiviral response activity that could have clinical importance in HIV infection.


Assuntos
Adjuvantes Imunológicos/biossíntese , Fármacos Anti-HIV/metabolismo , Antivirais/biossíntese , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Regulação para Cima/imunologia , Molécula 1 de Adesão de Célula Vascular/biossíntese , Doença Aguda , Adjuvantes Imunológicos/genética , Adjuvantes Imunológicos/fisiologia , Antivirais/genética , Antivirais/fisiologia , Linfócitos T CD8-Positivos/imunologia , Membrana Celular/imunologia , Membrana Celular/metabolismo , Membrana Celular/virologia , Células Cultivadas , Técnicas de Cocultura , Soronegatividade para HIV/imunologia , Soropositividade para HIV/genética , Soropositividade para HIV/imunologia , Soropositividade para HIV/metabolismo , HIV-1/crescimento & desenvolvimento , HIV-1/imunologia , Humanos , Interleucina-15/farmacologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/virologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , RNA Mensageiro/biossíntese , Linfócitos T/imunologia , Linfócitos T/virologia , Regulação para Cima/genética , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/fisiologia
10.
J Immunol ; 174(3): 1587-93, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15661920

RESUMO

Human CMV is often associated with transplant rejection and opportunistic infections such as pneumonia in immunosuppressed patients. Current anti-CMV therapies, although effective, show relatively high toxicity, which seriously limits their long-term use. In this study, we provide evidence that leukotriene B(4) (LTB(4)) plays an important role in the fight against murine CMV (MCMV) infection in vivo. Intravenous administration of 50 and 500 ng/kg/day of LTB(4) to mice infected with a lethal dose of MCMV significantly increases their survival (50 and 70%, respectively), compared with the placebo-treated group (10% of survival). In mice infected with a sublethal dose of MCMV and treated daily with 50 ng/kg/day of LTB(4), the salivary gland viral loads were found to be reduced by 66% compared with the control group. Furthermore, using an allogeneic bone marrow transplantation mouse model, the frequency of MCMV reactivation from latently infected mice was much lower (38%) in LTB(4) (500 ng/kg)-treated mice than in the placebo-treated group (78%). Finally, in experiments using 5-lipoxygenase-deficient mice, MCMV viral loads in salivary glands were found to be higher in animals unable to produce leukotrienes than in the control groups, supporting a role of endogenous 5-lipoxygenase products, possibly LTB(4), in host defense against CMV infection.


Assuntos
Antivirais/uso terapêutico , Transplante de Medula Óssea/efeitos adversos , Infecções por Citomegalovirus/prevenção & controle , Leucotrieno B4/uso terapêutico , Muromegalovirus/fisiologia , Ativação Viral , Latência Viral , Animais , Antivirais/deficiência , Antivirais/genética , Antivirais/fisiologia , Araquidonato 5-Lipoxigenase/deficiência , Araquidonato 5-Lipoxigenase/genética , Araquidonato 5-Lipoxigenase/fisiologia , Transplante de Medula Óssea/imunologia , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/mortalidade , Infecções por Citomegalovirus/virologia , Feminino , Rejeição de Enxerto/genética , Rejeição de Enxerto/prevenção & controle , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/prevenção & controle , Injeções Intravenosas , Leucotrieno B4/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Glândulas Salivares/efeitos dos fármacos , Glândulas Salivares/virologia , Baço/citologia , Baço/transplante , Baço/virologia , Carga Viral , Ativação Viral/genética , Ativação Viral/imunologia , Latência Viral/genética , Latência Viral/imunologia
11.
Curr Biol ; 15(2): 166-70, 2005 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-15668174

RESUMO

The antiretroviral activity of the cellular enzyme APOBEC3G has been attributed to the excessive deamination of cytidine (C) to uridine (U) in minus strand reverse transcripts, a process resulting in guanosine (G) to adenosine (A) hypermutation of plus strand DNAs. The HIV-1 Vif protein counteracts APOBEC3G by inducing proteasomal degradation and exclusion from virions through recruitment of a cullin5 ECS E3 ubiquitin ligase complex. APOBEC3G belongs to the APOBEC protein family, members of which possess consensus (H/C)-(A/V)-E-(X)24-30-P-C-(X)2-C cytidine deaminase motifs. Earlier analyses of APOBEC-1 have defined specific residues that are important for zinc coordination, proton transfer, and, therefore, catalysis within this motif. Because APOBEC3G contains two such motifs, we used site-directed mutagenesis of conserved residues to assess each region's contribution to anti-HIV-1 activity. Surprisingly, whereas either the N- or C-terminal domain could confer antiviral function in tissue culture-based infectivity assays, only an intact C-terminal motif was essential for DNA mutator activity. These findings reveal the nonequivalency of APOBEC3G's N- and C-terminal domains and imply that APOBEC3G-mediated DNA editing may not always be necessary for antiviral activity. Accordingly, we propose that APOBEC3G can achieve an anti-HIV-1 effect through an undescribed mechanism that is distinct from cytidine deamination.


Assuntos
Antivirais/fisiologia , Citidina Desaminase/metabolismo , Produtos do Gene vif/metabolismo , HIV-1 , Mutação/fisiologia , Proteínas/fisiologia , Desaminase APOBEC-3G , Motivos de Aminoácidos , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Humanos , Mutagênese Sítio-Dirigida , Mutação/genética , Nucleosídeo Desaminases , Estrutura Terciária de Proteína , Proteínas Repressoras , Vírion/metabolismo , Produtos do Gene vif do Vírus da Imunodeficiência Humana
12.
J Immunol ; 173(10): 6274-83, 2004 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-15528366

RESUMO

The aim of this study was to investigate the mechanism of HIV-1 neutralization using monocyte-derived macrophages (MDM) in comparison to PBMC as target cells. For this purpose, we analyzed neutralizing activities of different human polyclonal IgG samples purified from sera of HIV-1-infected individuals using a single cycle infection assay. We found an increase of the neutralizing titer when macrophages vs PBMC were used as target cells. Moreover, polyclonal IgG from HIV-1-infected patients that are not able to neutralize virus when PBMC are used as target cells strongly inhibit MDM infection. Similar results were obtained with neutralizing mAbs. To explore the participation of FcgammaRs in HIV-1 inhibition, F(ab')(2) and Fab of these Igs were produced. Results indicated that both F(ab')(2) and Fab are less effective to inhibit virus replication in MDM. Moreover, competition experiments with Fc fragments of IgG from healthy donors or with purified monoclonal anti-human FcgammaRs Ab strengthen the participation of the FcgammaRs, and in particular of FcgammaRI (CD64) in HIV-1 inhibition on MDM. Mechanisms by which HIV-specific IgG inhibit virus replication in cultured macrophages are proposed and the benefit of inducing such Abs by vaccination is discussed.


Assuntos
Antivirais/fisiologia , Infecções por HIV/imunologia , HIV-1/imunologia , Imunoglobulina G/isolamento & purificação , Imunoglobulina G/fisiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Receptores de IgG/fisiologia , Fármacos Anti-HIV/farmacologia , Ligação Competitiva/imunologia , Células Cultivadas , Quimiocinas/biossíntese , Humanos , Imunoglobulina A/isolamento & purificação , Imunoglobulina A/fisiologia , Fragmentos Fab das Imunoglobulinas/isolamento & purificação , Fragmentos Fab das Imunoglobulinas/fisiologia , Imunoglobulina G/biossíntese , Macrófagos/virologia , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/virologia , Testes de Neutralização , Fagocitose/imunologia , Ligação Proteica/imunologia , Receptores de IgG/imunologia , Receptores de IgG/metabolismo , Replicação Viral/imunologia
13.
J Immunol ; 173(10): 6303-11, 2004 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-15528369

RESUMO

A large proportion of the CD8(+) T cell pool in persons chronically infected with HIV consists of cells that show features of replicative senescence, an end stage characterized by irreversible cell cycle arrest, multiple genetic and functional changes, and shortened telomeres. The objective of our research was to determine whether constitutive expression of the gene for the human telomerase (hTERT) can prevent senescence-induced impairments in human virus-specific CD8(+) T cells, particularly in the context of HIV-1 disease. Our results indicate that hTERT-expressing HIV-specific CD8(+) lymphocytes show both an enhanced and sustained capacity to inhibit HIV-1 replication in in vitro coculture experiments, as well as prolonged ability to produce IFN-gamma and TNF-alpha in response to stimulation with HIV-1-derived peptides, as compared with vector-transduced controls. Loss of CD28 expression, the signature change of replicative senescence in cell culture, was retarded in those CD8(+) T cell cultures that had high levels of CD28 at the time of hTERT transduction. These findings suggest that telomere shortening may be the primary driving force behind several aspects of CD8(+) T cell dysfunction associated with replicative senescence. We also demonstrate reduced accumulation of the p16(INK4a) and p21(WAF1) cell cycle inhibitors in hTERT-transduced lymphocytes, providing a possible mechanism by which stable hTERT expression is able to circumvent the senescence barrier in CD8(+) T cells. Given the key role of CD8(+) T cell function in controlling a variety of acute and latent viral infections, approaches to retard the functional decrements associated with replicative senescence may lead to novel types of immunotherapy.


Assuntos
Adjuvantes Imunológicos/fisiologia , Antivirais/fisiologia , Linfócitos T CD8-Positivos/enzimologia , Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/imunologia , HIV-1/imunologia , Telomerase/fisiologia , Telômero/metabolismo , Adjuvantes Imunológicos/biossíntese , Adjuvantes Imunológicos/genética , Adjuvantes Imunológicos/metabolismo , Antivirais/biossíntese , Antivirais/genética , Antivirais/metabolismo , Antígenos CD28/biossíntese , Antígenos CD28/fisiologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/virologia , Ciclo Celular/genética , Ciclo Celular/imunologia , Proliferação de Células , Células Cultivadas , Senescência Celular/genética , Senescência Celular/imunologia , Citotoxicidade Imunológica/genética , Proteínas de Ligação a DNA , Inibidores do Crescimento/antagonistas & inibidores , Inibidores do Crescimento/biossíntese , HIV-1/crescimento & desenvolvimento , HIV-1/fisiologia , Humanos , Interferon gama/biossíntese , Interferon gama/fisiologia , Telomerase/biossíntese , Telomerase/genética , Telomerase/metabolismo , Telômero/enzimologia , Telômero/genética , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/fisiologia , Regulação para Cima/imunologia , Replicação Viral/genética , Replicação Viral/imunologia
14.
J Immunol ; 173(7): 4618-26, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15383596

RESUMO

During HIV-1 infection or vaccination, HIV-1 envelope spikes elicit Ab responses. Neutralizing Abs block viral entry by recognizing epitopes on spikes critical for their interaction with receptor, coreceptors or fusion. In contrast, nonneutralizing Abs fail to do so because they recognize epitopes either buried or exposed but not critical for viral entry. Previously, we produced a high-affinity human mAb against the cluster II determinant of gp41. This Ab or its recombinant Fab and single-chain Fv have been repeatedly shown to bind to HIV-1 gp160 or gp41, but fail to block viral entry. We report that, surprisingly, expression of this nonneutralizing anti-HIV-1 gp41 single-chain Fv on the surface of human CD4 T cells markedly inhibits HIV-1 replication and cell-cell fusion. The inhibition targets the HIV-1 envelope at the level of viral entry, regardless of HIV-1 tropism. Although this bona fide nonneutralizing Ab does not neutralize HIV-1 entry when produced as a soluble protein, it acts as a neutralizing Ab when expressed on the cell surface. Expressing Abs on the surface of HIV-1-susceptible cells can be a new way to fight HIV-1.


Assuntos
Antivirais/fisiologia , Anticorpos Anti-HIV/fisiologia , Infecções por HIV/imunologia , Infecções por HIV/prevenção & controle , HIV-1/imunologia , HIV-1/patogenicidade , Receptores de Antígenos de Linfócitos B/fisiologia , Antivirais/biossíntese , Linhagem Celular Tumoral , Membrana Celular/imunologia , Membrana Celular/virologia , Suscetibilidade a Doenças/imunologia , Produtos do Gene env/antagonistas & inibidores , Vetores Genéticos , Células Gigantes/imunologia , Células Gigantes/virologia , Proteínas de Fluorescência Verde , Anticorpos Anti-HIV/biossíntese , Inibidores da Fusão de HIV/imunologia , HIV-1/fisiologia , Humanos , Fragmentos de Imunoglobulinas/biossíntese , Fragmentos de Imunoglobulinas/genética , Fragmentos de Imunoglobulinas/fisiologia , Proteínas Luminescentes/genética , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/genética , Testes de Neutralização , Receptores de Antígenos de Linfócitos B/biossíntese , Transdução Genética , Transgenes , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/imunologia , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas do Envelope Viral/genética , Replicação Viral/imunologia
15.
J Immunol ; 173(6): 4108-19, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15356161

RESUMO

Adaptive cellular immunity is required to clear HSV-1 infection in the periphery. Myeloid dendritic cells (DCs) are the first professional Ag-presenting cell to encounter the virus after primary and secondary infection and thus the consequences of their infection are important in understanding the pathogenesis of the disease and the response to the virus. Following HSV-1 infection, both uninfected and infected human DCs acquire a more mature phenotype. In this study, we demonstrate that type I IFN secreted from myeloid DC mediates bystander activation of the uninfected DCs. Furthermore, we confirm that this IFN primes DCs for elevated IL-12 p40 and p70 secretion. However, secretion of IFN is not responsible for the acquisition of a mature phenotype by HSV-1-infected DC. Rather, virus binding to a receptor on the cell surface induces DC maturation directly, through activation of the NF-kappaB and p38 MAPK pathways. The binding of HSV glycoprotein D is critical to the acquisition of a mature phenotype and type I IFN secretion. The data therefore demonstrate that DCs can respond to HSV exposure directly through recognition of viral envelope structures. In the context of natural HSV infection, the coupling of viral entry to the activation of DC signaling pathways is likely to be counterbalanced by viral disruption of DC maturation. However, the parallel release of type I IFN may result in paracrine activation so that the DCs are nonetheless able to mount an adaptive immune response.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/virologia , Herpesvirus Humano 1/imunologia , Interferon Tipo I/metabolismo , Células Mieloides/imunologia , Células Mieloides/virologia , Comunicação Parácrina/imunologia , Antígenos CD/biossíntese , Antivirais/fisiologia , Antígeno B7-2 , Diferenciação Celular/imunologia , Membrana Celular/enzimologia , Membrana Celular/imunologia , Membrana Celular/metabolismo , Membrana Celular/virologia , Células Cultivadas , Células Dendríticas/metabolismo , Ativação Enzimática/imunologia , Herpesvirus Humano 1/fisiologia , Herpesvirus Humano 1/efeitos da radiação , Humanos , Interferon Tipo I/antagonistas & inibidores , Interferon Tipo I/fisiologia , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/biossíntese , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/virologia , Células Mieloides/metabolismo , NF-kappa B/metabolismo , Testes de Neutralização , Raios Ultravioleta , Regulação para Cima/imunologia , Proteínas do Envelope Viral/fisiologia , Inativação de Vírus/efeitos da radiação , Proteínas Quinases p38 Ativadas por Mitógeno
16.
Curr Biol ; 14(15): 1385-91, 2004 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-15296757

RESUMO

APOBEC3G (CEM15 ) deaminates cytosine to uracil in nascent retroviral cDNA. The potency of this cellular defense is evidenced by a dramatic reduction in viral infectivity and the occurrence of high frequencies of retroviral genomic-strand G --> A transition mutations. The overwhelming dinucleotide hypermutation preference of APOBEC3G acting upon a variety of model retroviral substrates is 5'-GG --> -AG. However, a distinct 5'-GA --> -AA bias, which is difficult to attribute to APOBEC3G alone, prevails in HIV-1 sequences derived from infected individuals (e.g., ). Here, we show that APOBEC3F is also a potent retroviral restrictor but that its activity, unlike that of APOBEC3G, is partially resistant to HIV-1 Vif and results in a clear 5'-GA --> -AA retroviral hypermutation preference. This bias is also apparent in a bacterial mutation assay, suggesting that it is an intrinsic APOBEC3F property. Moreover, APOBEC3F and APOBEC3G appear to be coordinately expressed in a wide range of human tissues and are independently able to inhibit retroviral infection. Thus, APOBEC3F and APOBEC3G are likely to function alongside one another in the provision of an innate immune defense, with APOBEC3F functioning as the major contributor to HIV-1 hypermutation in vivo.


Assuntos
Antivirais/fisiologia , Citosina Desaminase/metabolismo , HIV-1/genética , Mutagênese/genética , Proteínas/metabolismo , Desaminase APOBEC-3G , Sequência de Aminoácidos , Sequência de Bases , Northern Blotting , Citidina Desaminase , Primers do DNA , Genes vif/genética , HIV-1/metabolismo , Humanos , Dados de Sequência Molecular , Nucleosídeo Desaminases , Mutação Puntual/genética , Proteínas/genética , Proteínas Repressoras , Alinhamento de Sequência , Análise de Sequência de DNA , Replicação Viral/genética , Replicação Viral/fisiologia
17.
AIDS Res Hum Retroviruses ; 20(6): 600-7, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15242536

RESUMO

Human saliva contains multiple components that inhibit HIV-1 infection in vitro, which may contribute to low oral HIV-1 transmission. Salivary agglutinin (SAG) is a high-molecular-weight glycoprotein encoded by DMBT-1 and identical to gp340, a member of the lung scavange receptor, cysteine-rich receptor family. gp340 binds to surfactants A and D, which is believed to function in the clearance of microorganisms from the lung, as part of the innate immune response. Previously we reported that SAG (gp340) specifically inhibits HIV-1 infection with broad activity against diverse HIV-1 isolates. This gp340 inhibitory activity is mediated by binding to viral gp120 and involves a region different from the CD4-binding site on gp120. Here, we report that the gp340-binding region is localized to a linear, highly conserved sequence near the stem of the V3 loop that is critical for chemokine receptor interaction during viral binding and infection. The interaction of gp340 with gp120 is enhanced by prebinding of sCD4 to gp120, suggesting that gp340 inhibitory activity is mediated by blocking access of the gp120 to the chemokine receptor.


Assuntos
Aglutininas/metabolismo , Antivirais/fisiologia , Proteína gp120 do Envelope de HIV/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptores CCR5/metabolismo , Receptores de Superfície Celular/metabolismo , Antivirais/metabolismo , Sítios de Ligação , Antígenos CD4/metabolismo , Proteínas de Ligação ao Cálcio , Proteínas de Ligação a DNA , Humanos , Modelos Moleculares , Ligação Proteica , Proteínas Supressoras de Tumor
18.
J Virol ; 78(15): 8238-44, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15254195

RESUMO

Human APOBEC3G (huAPOBEC3G), also known as CEM15, is a broad antiretroviral host factor that deaminates dC to dU in the minus strand DNA of human immunodeficiency virus type 1 (HIV-1), other lentiviruses, and murine leukemia virus (MLV), thereby creating G-to-A hypermutation in the plus strand DNA to inhibit the infectivity of these viruses. In this study, we examined the antiretroviral function of a murine homologue of APOBEC3G (muAPOBEC3G) on several retrovirus systems with different producer cells. MuAPOBEC3G did not suppress the infectivity of murine retroviral vectors produced from human or murine cells, whereas it showed antiviral activity on both wild-type and Deltavif virions of HIV-1 in human cells. In contrast, huAPOBEC3G showed broad antiviral activity on HIV-1 and murine retroviral vectors produced from human cells as well as murine cells. These data suggested that muAPOBEC3G does not possess antiretroviral activity on murine retroviruses and has a different target specificity from that of huAPOBEC3G and that huAPOBEC3G works as a broad antiviral factor not only in human cells but also in murine cells. A functional interaction study between human and murine APOBEC3G supported the former hypothesis. Furthermore, studies on the expression of APOBEC3G in producer cells and its incorporation into virions revealed that muAPOBEC3G is incorporated into HIV-1 virions but not into MLV virions. Thus, muAPOBEC3G cannot suppress the infectivity of murine retrovirus because it is not incorporated into virions. We suggest that murine retroviruses can replicate in murine target cells expressing muAPOBEC3G because they are not targets for this enzyme.


Assuntos
Antivirais/fisiologia , Proteínas/fisiologia , Desaminase APOBEC-3G , Animais , Linhagem Celular , Citidina Desaminase , HIV-1/patogenicidade , Humanos , Vírus da Leucemia Murina/patogenicidade , Camundongos , Nucleosídeo Desaminases , Proteínas Repressoras , Especificidade da Espécie , Vírion/patogenicidade
19.
J Gen Virol ; 85(Pt 8): 2315-2326, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15269373

RESUMO

The interferon-induced murine Mx1 GTPase is a nuclear protein. It specifically inhibits influenza A viruses at the step of primary transcription, a process known to occur in the nucleus of infected cells. However, the exact mechanism of inhibition is still poorly understood. The Mx1 GTPase has previously been shown to accumulate in distinct nuclear dots that are spatially associated with promyelocytic leukaemia protein (PML) nuclear bodies (NBs), but the significance of this association is not known. Here it is reported that, in cells lacking PML and, as a consequence, PML NBs, Mx1 still formed nuclear dots. These dots were indistinguishable from the dots observed in wild-type cells, indicating that intact PML NBs are not required for Mx1 dot formation. Furthermore, Mx1 retained its antiviral activity against influenza A virus in these PML-deficient cells, which were fully permissive for influenza A virus. Nuclear Mx proteins from other species showed a similar subnuclear distribution. This was also the case for the human MxA GTPase when this otherwise cytoplasmic protein was translocated into the nucleus by virtue of a foreign nuclear localization signal. Human MxA and mouse Mx1 do not interact or form heterooligomers. Yet, they co-localized to a large degree when co-expressed in the nucleus. Taken together, these findings suggest that Mx1 dots represent distinct nuclear domains ('Mx nuclear domains') that are frequently associated with, but functionally independent of, PML NBs.


Assuntos
Antivirais/fisiologia , Proteínas de Ligação ao GTP/fisiologia , Vírus da Influenza A/crescimento & desenvolvimento , Proteínas de Neoplasias/fisiologia , Proteínas Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Animais , Núcleo Celular/química , Proteínas de Ligação ao GTP/análise , Camundongos , Proteínas de Resistência a Myxovirus , Proteína da Leucemia Promielocítica , Proteínas Supressoras de Tumor
20.
J Virol ; 78(11): 6073-6, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15141007

RESUMO

Recently, APOBEC3G has been identified as a host factor that blocks retroviral replication. It introduces G to A hypermutations in newly synthesized minus strand viral cDNA at the step of reverse transcription in target cells. Here, we identified the human APOBEC3F protein as another host factor that blocks human immunodeficiency virus type 1 (HIV-1) replication. Similar to APOBEC3G, APOBEC3F also induced G to A hypermutations in HIV genomic DNA, and the viral Vif protein counteracted its activity. Thus, APOBEC family members might have evolved as a general defense mechanism of the body against retroviruses, retrotransposons, and other mobile genetic elements.


Assuntos
Antivirais/fisiologia , Apolipoproteínas B/genética , Citidina Desaminase/fisiologia , HIV-1/fisiologia , Replicação Viral , Desaminase APOBEC-1 , Desaminase APOBEC-3G , Sequência de Aminoácidos , Sequência de Bases , Citidina Desaminase/química , Produtos do Gene vif/fisiologia , HIV-1/genética , Humanos , Dados de Sequência Molecular , Mutação , Nucleosídeo Desaminases , Proteínas/química , Proteínas Repressoras , Transcrição Gênica , Produtos do Gene vif do Vírus da Imunodeficiência Humana
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...