Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 7: 46719, 2017 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-28429755

RESUMO

APOBEC3s (A3s) are single-stranded DNA cytosine deaminases that provide innate immune defences against retroviruses and mobile elements. A3s are specific to eutherian mammals because no direct homologs exist at the syntenic genomic locus in metatherian (marsupial) or prototherian (monotreme) mammals. However, the A3s in these species have the likely evolutionary precursors, the antibody gene deaminase AID and the RNA/DNA editing enzyme APOBEC1 (A1). Here, we used cell culture-based assays to determine whether opossum A1 restricts the infectivity of retroviruses including human immunodeficiency virus type 1 (HIV-1) and the mobility of LTR/non-LTR retrotransposons. Opossum A1 partially inhibited HIV-1, as well as simian immunodeficiency virus (SIV), murine leukemia virus (MLV), and the retrotransposon MusD. The mechanism of inhibition required catalytic activity, except for human LINE1 (L1) restriction, which was deamination-independent. These results indicate that opossum A1 functions as an innate barrier to infection by retroviruses such as HIV-1, and controls LTR/non-LTR retrotransposition in marsupials.


Assuntos
Desaminase APOBEC-1/genética , Perfilação da Expressão Gênica , Gambás/genética , Retroelementos/genética , Retroviridae/genética , Desaminase APOBEC-1/metabolismo , Animais , DNA de Cadeia Simples/genética , Feminino , Células HEK293 , HIV-1/genética , Células HeLa , Humanos , Vírus da Leucemia Murina/genética , Masculino , Camundongos , Mutação , Gambás/metabolismo
2.
Sci Rep ; 6: 19035, 2016 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-26738439

RESUMO

APOBEC1 (A1) proteins from lagomorphs and rodents have deaminase-dependent restriction activity against HIV-1, whereas human A1 exerts a negligible effect. To investigate these differences in the restriction of HIV-1 by A1 proteins, a series of chimeric proteins combining rabbit and human A1s was constructed. Homology models of the A1s indicated that their activities derive from functional domains that likely act in tandem through a dimeric interface. The C-terminal region containing the leucine-rich motif and the dimerization domains of rabbit A1 is important for its anti-HIV-1 activity. The A1 chimeras with strong anti-HIV-1 activity were incorporated into virions more efficiently than those without anti-HIV-1 activity, and exhibited potent DNA-mutator activity. Therefore, the C-terminal region of rabbit A1 is involved in both its packaging into the HIV-1 virion and its deamination activity against both viral cDNA and genomic RNA. This study identifies the novel molecular mechanism underlying the target specificity of A1.


Assuntos
Desaminase APOBEC-1/farmacologia , Fármacos Anti-HIV/farmacologia , HIV-1/fisiologia , Proteínas Recombinantes de Fusão/farmacologia , Sequência de Aminoácidos , Animais , Avaliação Pré-Clínica de Medicamentos , Escherichia coli/genética , Genoma Bacteriano , Genoma Viral , Células HEK293 , HIV-1/efeitos dos fármacos , Humanos , Mutagênicos/farmacologia , Mutação , Multimerização Proteica , Coelhos , Homologia de Sequência de Aminoácidos , Vírion/efeitos dos fármacos , Vírion/fisiologia , Montagem de Vírus
3.
J Immunol ; 191(12): 6030-6039, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24198285

RESUMO

The ssDNA-dependent deoxycytidine deaminase apolipoprotein B mRNA-editing, enzyme-catalytic, polypeptide-like 3G (A3G) is a potent restrictive factor against HIV-1 virus lacking viral-encoded infectivity factor (Vif) in CD4(+) T cells. A3G antiretroviral activity requires its encapsulation into HIV-1 virions. In this study, we show that germinal center-associated nuclear protein (GANP) is induced in activated CD4(+) T cells and physically interacts with A3G. Overexpression of GANP augments the A3G encapsidation into the virion-like particles and ΔVif HIV-1 virions. GANP is encapsidated in HIV-1 virion and modulates A3G packaging into the cores together with cellular RNAs, including 7SL RNA, and with unspliced HIV-1 genomic RNA. GANP upregulation leads to a significant increase in A3G-catalyzed G→A hypermutation in the viral genome and suppression of HIV-1 infectivity in a single-round viral infection assay. Conversely, GANP knockdown caused a marked increase in HIV-1 infectivity in a multiple-round infection assay. The data suggest that GANP is a cellular factor that facilitates A3G encapsidation into HIV-1 virions to inhibit viral infectivity.


Assuntos
Acetiltransferases/fisiologia , Linfócitos T CD4-Positivos/imunologia , Citidina Desaminase/fisiologia , HIV-1/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Vírion/metabolismo , Desaminase APOBEC-3G , Acetiltransferases/antagonistas & inibidores , Acetiltransferases/biossíntese , Acetiltransferases/química , Acetiltransferases/genética , Células Cultivadas , Citidina Desaminase/química , Genes vif , HIV-1/ultraestrutura , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Ativação Linfocitária , Mutação , Mapeamento de Interação de Proteínas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Citoplasmático Pequeno/metabolismo , RNA Interferente Pequeno/farmacologia , RNA Viral/genética , RNA Viral/metabolismo , Partícula de Reconhecimento de Sinal/metabolismo , Regulação para Cima , Vírion/ultraestrutura , Virulência , Replicação Viral , Produtos do Gene vif do Vírus da Imunodeficiência Humana/deficiência , Pequeno RNA não Traduzido
4.
Front Microbiol ; 4: 28, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23431045

RESUMO

Over 40% of the human genome is recognizable as having been derived from ancient retroelements, transported by an intracellular copy-and-paste process involving an RNA intermediate, with an additional few percent classified as DNA transposable elements. Endogenous retroviruses are long terminal repeat (LTR)-type retroelements that account for ~8% of human genomic DNA. Non-LTR members are present at extremely high copy numbers, with ~17% of the human genome consisting of long interspersed nuclear elements (LINEs). These LINEs modify vertebrate genomes not only through insertions, but also by the indirect replication of non-autonomous retrotransposons, such as short interspersed nuclear elements. As expected, vertebrate intrinsic immunity has evolved to support a balance between retroelement insertions that confer beneficial genetic diversity and those that cause deleterious gene disruptions. The mammalian cytidine deaminases encoded by the APOBEC3 genes can restrict a broad number of exogenous pathogens, such as exogenous retroviruses, and the mobility of endogenous retroelements. Furthermore, APOBEC1 from a variety of mammalian species, which mediates the cytidine (C) to uridine (U) deamination of apolipoprotein B (apoB) mRNA, a protein involved in lipid transport, also plays a role in controlling mobile elements. These mammalian apoB mRNA-editing, catalytic polypeptide (APOBEC) cytidine deaminases, which can bind to single-stranded DNA (ssDNA) as well as RNA, are able to insert mutations into ssDNA and/or RNA as a result of their ability to deaminate C to U. While these APOBEC cytidine deaminases with DNA mutagenic activity can be deleterious to cells, their biological modifications, such as protein-protein interactions and subcellular localization, in addition to their ability to bind to RNA, appear to have conferred a role for APOBECs as a cellular defense system against retroviruses and retroelements. In support of this notion, the expansion of the single APOBEC3 gene in mice to the seven APOBEC3 genes found in primates apparently correlates with the significant enhancement of the restriction of endogenous retroelements seen in primates, including humans. This review discusses the current understanding of the mechanism of action of APOBEC cytidine deaminases and attempts to summarize their roles in controlling retrotransposons.

5.
Wiley Interdiscip Rev RNA ; 3(4): 529-41, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22549984

RESUMO

Apolipoprotein B (apo B) messenger RNA (mRNA)-editing, catalytic polypeptide (APOBEC) cytidine deaminases (CDAs), which can insert mutations into DNA and/or RNA as a result of their ability to deaminate cytidine (C) to uridine (U), originated from a branch of the zinc-dependent deaminase superfamily at the beginning of vertebrate radiation. The ability of mammalian CDAs encoded by the APOBEC3 genes to restrict a broad number of endogenous retroelements and exogenous retroviruses, including human immunodeficiency virus-1, is well established. Furthermore, APOBEC1 from a variety of mammalian species, which mediates the C-to-U deamination of apo B mRNA, a protein involved in lipid transport, also has a role in controlling mobile elements. A large portion of the mammalian genome is derived from ancient transposable elements. Retroelements, transported by an intracellular copy-and-paste process involving an RNA intermediate, constitute the majority of these mobile genetic elements. Endogenous retroviruses are long-terminal repeat (LTR)-type retroelements that account for approximately 10% of human and murine genomic DNA. Non-LTR members are present in extremely high copy numbers, with approximately 40% of the human and murine genomes consisting of long-interspersed nuclear element-1 (L1). These L1 elements modify mammalian genomes not only through insertions but also by the indirect replication of non-autonomous retrotransposons. As expected, vertebrate intrinsic immunity has evolved to support a balance between retroelement insertions that cause deleterious gene disruptions and those that confer beneficial genetic diversity. This review discusses the current understanding of the mechanism of action of APOBEC CDAs and their role in controlling retroviruses and retroelements.


Assuntos
Citidina Desaminase/metabolismo , Retroviridae/metabolismo , Desaminase APOBEC-1 , Animais , Humanos , Retroelementos
6.
J Biol Chem ; 286(16): 14049-56, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21357691

RESUMO

The human immunodeficiency virus 1 (HIV-1) viral protein R (Vpr) is an accessory protein that has been shown to have multiple roles in HIV-1 pathogenesis. By screening chemical libraries in the RIKEN Natural Products Depository, we identified a 3-phenyl coumarin-based compound that inhibited the cell cycle arrest activity of Vpr in yeast and Vpr-dependent viral infection of human macrophages. We determined its minimal pharmacophore through a structure-activity relationship study and produced more potent derivatives. We detected direct binding, and by assaying a panel of Vpr mutants, we found the hydrophobic region about residues Glu-25 and Gln-65 to be potentially involved in the binding of the inhibitor. Our findings exposed a targeting site on Vpr and delineated a convenient approach to explore other targeting sites on the protein using small molecule inhibitors as bioprobes.


Assuntos
Fármacos Anti-HIV/farmacologia , Carbamatos/farmacologia , Cumarínicos/química , Cumarínicos/farmacologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/antagonistas & inibidores , Sítios de Ligação , Ligação Competitiva , Relação Dose-Resposta a Droga , Desenho de Fármacos , Ácido Glutâmico/química , Glutamina/química , Humanos , Macrófagos/citologia , Mutação , Ligação Proteica , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/química
7.
Nucleic Acids Res ; 39(13): 5538-54, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21398638

RESUMO

The ability of mammalian cytidine deaminases encoded by the APOBEC3 (A3) genes to restrict a broad number of endogenous retroelements and exogenous retroviruses, including murine leukemia virus and human immunodeficiency virus (HIV)-1, is now well established. The RNA editing family member apolipoprotein B (apo B)-editing catalytic subunit 1 (APOBEC1; A1) from a variety of mammalian species, a protein involved in lipid transport and which mediates C-U deamination of mRNA for apo B, has also been shown to modify a range of exogenous retroviruses, but its activity against endogenous retroelements remains unclear. Here, we show in cell culture-based retrotransposition assays that A1 family proteins from multiple mammalian species can also reduce the mobility and infectivity potential of LINE-1 (long interspersed nucleotide sequence-1, L1) and long-terminal repeats (LTRs) retrotransposons (or endogenous retroviruses), such as murine intracisternal A-particle (IAP) and MusD sequences. The anti-L1 activity of A1 was mainly mediated by a deamination-independent mechanism, and was not affected by subcellular localization of the proteins. In contrast, the inhibition of LTR-retrotransposons appeared to require the deaminase activity of A1 proteins. Thus, the AID/APOBEC family proteins including A1s employ multiple mechanisms to regulate the mobility of autonomous retrotransposons in several mammalian species.


Assuntos
Citidina Desaminase/metabolismo , Retroelementos , Desaminase APOBEC-1 , Sequência de Aminoácidos , Animais , Bactérias/genética , Linhagem Celular , Citidina Desaminase/química , Citidina Desaminase/genética , DNA/biossíntese , Genes de Partícula A Intracisternal , Humanos , Elementos Nucleotídeos Longos e Dispersos , Camundongos , Dados de Sequência Molecular , Mutação , RNA/metabolismo , Coelhos , Ratos , Sequências Repetidas Terminais
8.
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
9.
Nucleic Acids Res ; 36(21): 6859-71, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18971252

RESUMO

Although the role of the APOBEC3-dependent retroelement restriction system as an intrinsic immune defense against human immunodeficiency virus type1 (HIV-1) infection is becoming clear, only the rat ortholog of mammalian APOBEC1s (A1) thus far has been shown to possess antiviral activity. Here, we cloned A1 cDNAs from small animal species, and showed that similar to rat A1, both wild-type and Deltavif HIV-1 infection was inhibited by mouse and hamster A1 (4- to 10-fold), whereas human A1 had negligible effects. Moreover, rabbit A1 significantly reduced the infectivity of both HIV-1 virions (>300-fold), as well as that of SIVmac, SIVagm, FIV and murine leukemia virus. Immunoblot analysis showed that A1s were efficiently incorporated into the HIV-1 virion, and their packaging is mediated through an interaction with the nucleocapsid Gag domain. Interestingly, there was a clear accumulation of particular C-T changes in the genomic RNAs of HIV-1 produced in their presence, with few G-A changes in the proviral DNA. Together, these data reveal that A1 may function as a defense mechanism, regulating retroelements in a wide range of mammalian species.


Assuntos
Citidina Desaminase/metabolismo , HIV-1/fisiologia , Lentivirus/fisiologia , Desaminase APOBEC-1 , Sequência de Aminoácidos , Animais , Linhagem Celular , Clonagem Molecular , Cricetinae , Citidina Desaminase/classificação , Citidina Desaminase/genética , DNA Viral/química , DNA Viral/metabolismo , HIV-1/genética , HIV-1/metabolismo , Humanos , Vírus da Leucemia Murina/fisiologia , Camundongos , Dados de Sequência Molecular , Filogenia , Estrutura Terciária de Proteína , RNA Viral/química , RNA Viral/metabolismo , Coelhos , Ratos , Vírion/fisiologia , Montagem de Vírus , Produtos do Gene gag do Vírus da Imunodeficiência Humana/química
10.
Int Rev Immunol ; 27(4): 225-53, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18574738

RESUMO

Human T-cell leukemia virus type I (HTLV-I) is the first human retrovirus and causes adult T-cell leukemia/lymphoma (ATL). Constitutive activation of nuclear factor-kappa B (NF-kappa B) in the leukemic cells is essential for their growth and survival. Thus, NF-kappa B inhibitors have been attracting attention as a potential strategy to treat ATL. Recently, the field of retrovirus research has been stimulated by the discovery of an innate host defense factor, APOBEC3, against the retroviruses. HTLV-I is relatively resistant to the antiviral effects of APOBEC3. To clarify the resistance of HTLV-I against APOBEC3 might contribute to the design of effective therapeutic approaches.


Assuntos
Citosina Desaminase/farmacologia , Infecções por HTLV-I/prevenção & controle , Vírus Linfotrópico T Tipo 1 Humano/efeitos dos fármacos , Proteínas I-kappa B/farmacologia , Leucemia-Linfoma de Células T do Adulto/prevenção & controle , NF-kappa B/antagonistas & inibidores , Inibidores da Transcriptase Reversa/farmacologia , Desaminases APOBEC , Adulto , Animais , Citidina Desaminase , Citosina Desaminase/imunologia , Infecções por HTLV-I/imunologia , Infecções por HTLV-I/metabolismo , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Humanos , Proteínas I-kappa B/imunologia , Imunidade Inata , Leucemia-Linfoma de Células T do Adulto/imunologia , Leucemia-Linfoma de Células T do Adulto/metabolismo , Inibidor de NF-kappaB alfa , NF-kappa B/imunologia , Inibidores da Transcriptase Reversa/imunologia , Replicação Viral/efeitos dos fármacos
11.
J Virol Methods ; 139(1): 93-6, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17123636

RESUMO

The objective of this study was to investigate whether the anti-retroviral cellular cytidine deaminase, APOBEC3, inhibits the infectivity of human T cell leukemia virus type I (HTLV-I). Sufficient quantities of cell-free HTLV-I virion for infection were obtained by cotransfecting cells with HTLV-I and human or murine APOBEC3 expression vectors along with a plasmid expressing Tax. HTLV-I viruses containing these deaminases were still capable of infecting 293T and MOLT-4 cells. No G-to-A mutations, which are characteristic of cytidine deaminases, were observed in the HTLV-I genome. These results suggest that the enzymatic activity of APOBEC3 may not contribute substantially to antiviral responses to HTLV-I.


Assuntos
Antivirais/farmacologia , Citidina Desaminase/farmacologia , Citosina Desaminase/farmacologia , Vírus Linfotrópico T Tipo 1 Humano/efeitos dos fármacos , Desaminases APOBEC , Animais , Linhagem Celular , Farmacorresistência Viral , Transfecção
12.
FEBS Lett ; 580(11): 2598-602, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16631749

RESUMO

HIV-1 viral protein R (Vpr) is one of the human immunodeficiency virus type 1 encoded proteins that have important roles in viral pathogenesis. However, no clinical drug for AIDS therapy that targets Vpr has been developed. Here, we have established a screening system to isolate Vpr inhibitors using budding yeast cells. We purified a Vpr inhibitory compound from fungal metabolites and identified it as fumagillin, a chemical already known to be a potent inhibitor of angiogenesis. Fumagillin not only reversed the growth inhibitory activity of Vpr in yeast and human cells, but also inhibited Vpr-dependent viral gene expression upon the infection of human macrophages.


Assuntos
Ácidos Graxos Insaturados/farmacologia , Produtos do Gene vpr/antagonistas & inibidores , HIV-1/efeitos dos fármacos , HIV-1/fisiologia , Macrófagos/efeitos dos fármacos , Macrófagos/virologia , Aminopeptidases/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células , Cicloexanos , Avaliação Pré-Clínica de Medicamentos , Ácidos Graxos Insaturados/química , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Produtos do Gene vpr/metabolismo , Humanos , Metaloendopeptidases/metabolismo , Estrutura Molecular , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/efeitos dos fármacos , Sesquiterpenos , Transdução de Sinais/efeitos dos fármacos , Produtos do Gene vpr do Vírus da Imunodeficiência Humana
13.
J Virol ; 77(9): 5109-17, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12692213

RESUMO

In vivo studies for understanding viral transmission and replication, host immune responses, and pathogenesis of human immunodeficiency virus type 1 (HIV-1) infection would greatly benefit from the establishment of a small-animal model. In this study, we explored the potential of American mink (Mustera vison) as a susceptible host. We found that primary cells and cell lines derived from this species efficiently supported trans-activation of the HIV-1 long terminal repeat by Tat. Accordingly, the cysteine residue at position 261, which has been shown to be important for interaction of the human cyclin T1 with the HIV-1 regulatory protein Tat, is conserved in the mink homologue. No species-specific defect in Rev function could be detected in mink cells. In addition, primary splenocytes, fibroblasts, and the Mv.1.Lu cell line from American mink supported early as well as late HIV-1 gene expression following infection with vesicular stomatitis G protein-pseudotyped HIV-1 viruses, at levels comparable to those seen with permissive human cells. Furthermore, the mink Mv.1.Lu cell line stably expressing human CD4 and CCR5 receptors supported a spreading HIV-1 infection with few, if any, deficiencies compared to findings in human cell lines. This indicates the potential of HIV-1 to replicate in these cells once the blockade at the stage of virus entry has been removed. These results clearly show that cells from American mink generally pose no functional intracellular block to HIV-1 replication, and collectively they raise the possibility that this animal species could be engineered to support HIV-1 infection, providing a useful small-animal model for evaluating de novo infection by HIV-1.


Assuntos
Modelos Animais de Doenças , Fibroblastos/virologia , HIV-1/fisiologia , Vison/virologia , Baço/virologia , Replicação Viral , Sequência de Aminoácidos , Animais , Linhagem Celular , Células Cultivadas , Ciclina T , Ciclinas/metabolismo , Fibroblastos/citologia , Expressão Gênica , Produtos do Gene tat/metabolismo , Infecções por HIV/virologia , Repetição Terminal Longa de HIV/genética , HIV-1/genética , Humanos , Camundongos , Dados de Sequência Molecular , Baço/citologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana
14.
Virology ; 305(1): 181-91, 2003 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-12504551

RESUMO

We examine the potential for a broad range of small animal cells, including rodent, mink, and avian cells, from multiple tissues to support postintegration steps of HIV-1 replication. These cells were engineered so as to support a stable expression of human cyclin T1 and were further transduced with HIV-1 gag and pol genes. Viral gene expression was activated by the presence of human cyclin T1, but, with the exception of mink cells, was not at the level seen in human cells. Furthermore, there were considerable defects in p24 CA release, in particular in the case of rodent cells. Fractionation of Gag proteins by sucrose floatation revealed that the Gag in human cells trafficked to membrane fractions and was processed to p24 CA and p17 MA efficiently. Confocal imaging demonstrated that Gag was localized in a punctate pattern at the plasma membrane as well as intracellular membrane trans-Golgi cisternae in these cells. In contrast, the majority of Gag in rodent cells was largely present in cytosolic complexes and remained unprocessed. Labeling with [9,10(n)-(3)H]myristic acid showed a similar degree of N-myristoylated Pr55(gag) in rodent and human cells, indicating that while N-myristoylation of Gag was essential for membrane binding, it was not sufficient to confer membrane targeting specificity. Remarkably, despite the reduced level of intracellular Gag processing, mink Mv.1.Lu cells did not appear to differ significantly from human cells in support of virion assembly and release. Analysis of reciprocal heterokaryons suggested that the cellular factor(s) required for efficient assembly and release of infectious virions is lacking in murine cells but appears to be functionally present in mink as well as human cells. Our findings confirm and extend previous reports of multiple blocks to HIV replication in nonhuman cells that are most profound in murine cells. They also raise the possibility that other small animals, such as mink, could serve as novel model systems for studying HIV-1 infection and disease.


Assuntos
Produtos do Gene gag/fisiologia , HIV-1/fisiologia , Integração Viral , Replicação Viral , Células 3T3 , Animais , Linhagem Celular , Ciclina T , Ciclinas/fisiologia , Produtos do Gene gag/análise , Produtos do Gene gag/metabolismo , Produtos do Gene pol/fisiologia , Humanos , Camundongos , Vison , Ácido Mirístico/metabolismo , Precursores de Proteínas/metabolismo , Vírion/fisiologia , Montagem de Vírus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA