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











Base de dados
Intervalo de ano de publicação
1.
Inflamm Res ; 69(7): 683-696, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32347316

RESUMO

OBJECTIVE: Hepatitis B virus X protein (HBx) is a pivotal factor for HBV-induced hepatitis. Herein, we sought to investigate HBx-mediated NLR pyrin domain containing 3 (NLRP3) inflammasome activation and pyroptosis under oxidative stress. METHODS: The effect of HBx on the NLRP3 inflammasome was analyzed by enzyme-linked immunosorbent assays, quantitative reverse transcription-polymerase chain reaction, western blotting, and immunofluorescence in hepatic HL7702 cells. Pyroptosis was evaluated by western blotting, lactate dehydrogenase release, propidium iodide staining, and transmission electron microscopy. NLRP3 expression in the inflammasome from liver tissues was assessed by immunohistochemistry. RESULTS: In hydrogen peroxide (H2O2)-stimulated HL7702 cells, HBx triggered the release of pro-inflammatory mediators apoptosis-associated speck-like protein containing a CARD (ASC), interleukin (IL)-1ß, IL-18, and high-mobility group box 1 (HMGB1); activated NLRP3; and initiated pro-inflammatory cell death (pyroptosis). HBx localized to the mitochondria, where it induced mitochondrial damage and production of mitochondrial reactive oxygen species (mitoROS). Treatment of HL7702 cells with a mitoROS scavenger attenuated HBx-induced NLRP3 activation and pyroptosis. Expression levels of NLRP3, ASC, and IL-1ß in liver tissues from patients were positively correlated with HBV DNA concentration. CONCLUSIONS: The NLRP3 inflammasome was activated by elevated mitoROS levels and mediated HBx-induced liver inflammation and hepatocellular pyroptosis under H2O2-stress conditions.


Assuntos
Hepatócitos/patologia , Inflamassomos/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Estresse Oxidativo , Piroptose/efeitos dos fármacos , Transativadores/farmacologia , Proteínas Virais Reguladoras e Acessórias/farmacologia , Proteínas Adaptadoras de Sinalização CARD/sangue , Carcinoma Hepatocelular/virologia , Linhagem Celular , DNA Viral/análise , Expressão Gênica , Vírus da Hepatite B/genética , Hepatócitos/metabolismo , Humanos , Peróxido de Hidrogênio/farmacologia , Neoplasias Hepáticas/virologia , Mitocôndrias Hepáticas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transativadores/genética , Transfecção , Proteínas Virais Reguladoras e Acessórias/genética
2.
Retrovirology ; 15(1): 69, 2018 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-30316304

RESUMO

BACKGROUND: SAM domain and HD domain containing protein 1 (SAMHD1) is a host anti-HIV-1 restriction factor known to suppress viral reverse transcription in nondividing myeloid cells by its dNTP triphosphorylase activity that depletes cellular dNTPs. However, HIV-2 and some SIV strains rapidly replicate in macrophages due to their accessory protein, viral protein X (Vpx), which proteosomally degrades SAMHD1 and elevates dNTP levels. Endogenous reverse transcription (ERT) of retroviruses is the extra-cellular reverse transcription step that partially synthesizes proviral DNAs within cell-free viral particles before the viruses infect new cells. ERT activity utilizes dNTPs co-packaged during budding from the virus-producing cells, and high ERT activity is known to enhance HIV-1 infectivity in nondividing cells. Here, since Vpx elevates cellular dNTP levels in macrophages, we hypothesize that HIV-2 should contain higher ERT activity than HIV-1 in macrophages, and that the Vpx-mediated dNTP elevation should enhance both ERT activity and infectivity of HIV-1 particles produced in macrophages. RESULTS: Here, we demonstrate that HIV-2 produced from human primary monocyte derived macrophages displays higher ERT activity than HIV-1 produced from macrophages. Also, HIV-1 particles produced from macrophages treated with virus like particles (VLPs) containing Vpx, Vpx (+), displayed large increases of ERT activity with the enhanced copy numbers of early, middle and late reverse transcription products within the viral particles, compared to the viruses produced from macrophages treated with Vpx (-) VLPs. Furthermore, upon the infection with an equal p24 amount to fresh macrophages, the viruses produced from the Vpx (+) VLP treated macrophages demonstrated higher infectivity than the viruses from the Vpx (-) VLP treated macrophages. CONCLUSIONS: This finding identifies the viral ERT step as an additional step of HIV-1 replication cycle that SAMHD1 restricts in nondividing myeloid target cells.


Assuntos
HIV-1/genética , HIV-2/genética , Macrófagos/virologia , Transcrição Reversa/genética , Proteína 1 com Domínio SAM e Domínio HD/genética , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/virologia , Células Cultivadas , Citoplasma/química , Desoxirribonucleotídeos/análise , HIV-1/fisiologia , HIV-2/fisiologia , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/farmacologia , Vírion , Replicação Viral
3.
Cell Host Microbe ; 23(5): 607-617.e6, 2018 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-29746832

RESUMO

Bacteriophage-encoded genetic elements control bacterial biological functions. Enterohemorrhagic Escherichia coli (EHEC) strains harbor lambda-phages encoding the Shiga-toxin (Stx), which is expressed during the phage lytic cycle and associated with exacerbated disease. Phages also reside dormant within bacterial chromosomes through their lysogenic cycle, but how this impacts EHEC virulence remains unknown. We find that during lysogeny the phage transcription factor Cro activates the EHEC type III secretion system (T3SS). EHEC lambdoid phages are lysogenic under anaerobic conditions when Cro binds to and activates the promoters of T3SS genes. Interestingly, the Cro sequence varies among phages carried by different EHEC outbreak strains, and these changes affect Cro-dependent T3SS regulation. Additionally, infecting mice with the related pathogen C. rodentium harboring the bacteriophage cro from EHEC results in greater T3SS gene expression and enhanced virulence. Collectively, these findings reveal the role of phages in impacting EHEC virulence and their potential to affect outbreak strains.


Assuntos
Colífagos/metabolismo , Escherichia coli Êntero-Hemorrágica/efeitos dos fármacos , Escherichia coli Êntero-Hemorrágica/genética , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Proteínas Repressoras/farmacologia , Proteínas Virais Reguladoras e Acessórias/farmacologia , Fatores de Virulência/genética , Animais , Cromossomos Bacterianos/efeitos dos fármacos , Citrobacter rodentium/patogenicidade , Colífagos/genética , Modelos Animais de Doenças , Infecções por Enterobacteriaceae/metabolismo , Infecções por Enterobacteriaceae/patologia , Escherichia coli Êntero-Hemorrágica/patogenicidade , Escherichia coli Êntero-Hemorrágica/virologia , Escherichia coli/genética , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Genes Bacterianos/efeitos dos fármacos , Células HeLa , Humanos , Lipídeos , Lisogenia , Camundongos , Camundongos Endogâmicos C3H , Proteínas Repressoras/genética , Toxina Shiga/genética , Fatores de Transcrição , Sistemas de Secreção Tipo III/efeitos dos fármacos , Sistemas de Secreção Tipo III/genética , Sistemas de Secreção Tipo III/metabolismo , Ensaio de Placa Viral , Proteínas Virais Reguladoras e Acessórias/genética , Virulência/efeitos dos fármacos , Virulência/genética
4.
Nat Med ; 23(2): 256-263, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28067901

RESUMO

The cytostatic deoxycytidine analog cytarabine (ara-C) is the most active agent available against acute myelogenous leukemia (AML). Together with anthracyclines, ara-C forms the backbone of AML treatment for children and adults. In AML, both the cytotoxicity of ara-C in vitro and the clinical response to ara-C therapy are correlated with the ability of AML blasts to accumulate the active metabolite ara-C triphosphate (ara-CTP), which causes DNA damage through perturbation of DNA synthesis. Differences in expression levels of known transporters or metabolic enzymes relevant to ara-C only partially account for patient-specific differential ara-CTP accumulation in AML blasts and response to ara-C treatment. Here we demonstrate that the deoxynucleoside triphosphate (dNTP) triphosphohydrolase SAM domain and HD domain 1 (SAMHD1) promotes the detoxification of intracellular ara-CTP pools. Recombinant SAMHD1 exhibited ara-CTPase activity in vitro, and cells in which SAMHD1 expression was transiently reduced by treatment with the simian immunodeficiency virus (SIV) protein Vpx were dramatically more sensitive to ara-C-induced cytotoxicity. CRISPR-Cas9-mediated disruption of the gene encoding SAMHD1 sensitized cells to ara-C, and this sensitivity could be abrogated by ectopic expression of wild-type (WT), but not dNTPase-deficient, SAMHD1. Mouse models of AML lacking SAMHD1 were hypersensitive to ara-C, and treatment ex vivo with Vpx sensitized primary patient-derived AML blasts to ara-C. Finally, we identified SAMHD1 as a risk factor in cohorts of both pediatric and adult patients with de novo AML who received ara-C treatment. Thus, SAMHD1 expression levels dictate patient sensitivity to ara-C, providing proof-of-concept that the targeting of SAMHD1 by Vpx could be an attractive therapeutic strategy for potentiating ara-C efficacy in hematological malignancies.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Citarabina/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Proteínas Monoméricas de Ligação ao GTP/efeitos dos fármacos , Proteínas Virais Reguladoras e Acessórias/farmacologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antimetabólitos Antineoplásicos/uso terapêutico , Arabinofuranosilcitosina Trifosfato/metabolismo , Criança , Pré-Escolar , Citarabina/uso terapêutico , Modelos Animais de Doenças , Feminino , Humanos , Técnicas In Vitro , Lactente , Leucemia Mieloide Aguda/metabolismo , Masculino , Camundongos , Terapia de Alvo Molecular , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Prognóstico , Proteína 1 com Domínio SAM e Domínio HD
5.
Sci Rep ; 6: 38162, 2016 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-27922067

RESUMO

SAMHD1 restricts human immunodeficiency virus type 1 (HIV-1) replication in myeloid cells and CD4+ T cells, while Vpx can mediate SAMHD1 degradation to promote HIV-1 replication. Although the restriction mechanisms of SAMHD1 have been well-described, SAMHD1 expression and Vpx-mediated SAMHD1 degradation during chronic HIV-1 infection were poorly understood. Flow cytometric analysis was used to directly visualize ex vivo, and after in vitro SIV-Vpx treatment, SAMHD1 expression in CD4+ T cells and monocytes. Here we report activated CD4+ T cells without SAMHD1 expression were severely reduced, and SAMHD1 in CD4+ T cells became susceptible to SIV-Vpx mediated degradation during chronic HIV-1 infection, which was absent from uninfected donors. These alterations were irreversible, even after long-term fully suppressive antiretroviral treatment. Although SAMHD1 expression in CD4+ T cells and monocytes was not found to correlate with plasma viral load, Vpx-mediated SAMHD1 degradation was associated with indicators of immune activation. In vitro assays further revealed that T-cell activation and an upregulated IFN-I pathway contributed to these altered SAMHD1 properties. These findings provide insight into how immune activation during HIV-1 infection leads to irreparable aberrations in restriction factors and in subsequent viral evasion from host antiviral defenses.


Assuntos
Infecções por HIV/imunologia , Proteína 1 com Domínio SAM e Domínio HD/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Adulto , Idoso , Fármacos Anti-HIV/uso terapêutico , Linhagem Celular , Doença Crônica , Feminino , Infecções por HIV/tratamento farmacológico , Infecções por HIV/metabolismo , HIV-1/patogenicidade , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Interferon gama/metabolismo , Masculino , Pessoa de Meia-Idade , Proteólise , Proteína 1 com Domínio SAM e Domínio HD/genética , Proteína 1 com Domínio SAM e Domínio HD/imunologia , Carga Viral , Proteínas Virais Reguladoras e Acessórias/farmacologia , Replicação Viral
6.
PLoS One ; 10(10): e0140561, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26496699

RESUMO

Two cellular factors are currently known to modulate lentiviral infection specifically in myeloid cells: SAMHD1 and APOBEC3A (A3A). SAMHD1 is a deoxynucleoside triphosphohydrolase that interferes with viral infection mostly by limiting the intracellular concentrations of dNTPs, while A3A is a cytidine deaminase that has been described to edit incoming vDNA. The restrictive phenotype of myeloid cells can be alleviated through the direct degradation of SAMHD1 by the HIV-2/SIVSM Vpx protein or else, at least in the case of HIV-1, by the exogenous supplementation of nucleosides that artificially overcome the catabolic activity of SAMHD1 on dNTPs. Here, we have used Vpx and dNs to explore the relationship existing between vDNA cytidine deamination and SAMHD1 during HIV-1 or SIVMAC infection of primary dendritic cells. Our results reveal an interesting inverse correlation between conditions that promote efficient infection of DCs and the extent of vDNA editing that may reflect the different susceptibility of vDNA to cytoplasmic effectors during the infection of myeloid cells.


Assuntos
Citidina/genética , DNA Viral/genética , Células Dendríticas/virologia , HIV-1/fisiologia , Nucleosídeos/farmacologia , Vírus da Imunodeficiência Símia/fisiologia , Proteínas Virais Reguladoras e Acessórias/farmacologia , Células Dendríticas/efeitos dos fármacos , Células HEK293 , HIV-1/efeitos dos fármacos , Células HeLa , Humanos , Cinética , Transcrição Reversa/efeitos dos fármacos , Vírus da Imunodeficiência Símia/efeitos dos fármacos
7.
Retrovirology ; 11: 63, 2014 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-25158827

RESUMO

BACKGROUND: SAMHD1 degrades deoxyribonucleotides (dNTPs), suppressing viral DNA synthesis in macrophages. Recently, viral protein X (Vpx) of HIV-2/SIVsm was shown to target SAMHD1 for proteosomal degradation and led to elevation of dNTP levels, which in turn accelerated proviral DNA synthesis of lentiviruses in macrophages. RESULTS: We investigated both time-dependent and quantitative interplays between SAMHD1 level and dNTP concentrations during multiple exposures of Vpx in macrophages. The following were observed. First, SAMHD1 level was rapidly reduced by Vpx + VLP to undetectable levels by Western blot analysis. Recovery of SAMHD1 was very slow with less than 3% of the normal macrophage level detected at day 6 post Vpx treatment and only ~30% recovered at day 14. Second, dGTP, dCTP and dTTP levels peaked at day 1 post Vpx treatment, whereas dATP peaked at day 2. However, all dNTPs rapidly decreased starting at day 3, while SAMHD1 level was below the level of detection. Third, when Vpx pretreated macrophages were re-exposed to a second Vpx treatment at day 7, we observed dNTP elevation that had faster kinetics than the first Vpx + VLP treatment. Moreover, we performed a short kinetic analysis of the second Vpx treatment to find that dATP and dGTP levels peaked at 8 hours post secondary VLP treatment. dGTP peak was consistently higher than the primary, whereas peak dATP concentration was basically equivalent to the first Vpx + VLP treatment. Lastly, HIV-1 replication kinetics were faster in macrophages treated after the secondary Vpx treatments when compared to the initial single Vpx treatment. CONCLUSION: This study reveals that a very low level of SAMHD1 sufficiently modulates the normally low dNTP levels in macrophages and proposes potential diverse mechanisms of Vpx-mediated dNTP regulation in macrophages.


Assuntos
Macrófagos/metabolismo , Monócitos/citologia , Proteínas Monoméricas de Ligação ao GTP/análise , Nucleotídeos/análise , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , HIV-1/genética , Humanos , Proteína 1 com Domínio SAM e Domínio HD , Espectrometria de Massas em Tandem , Proteínas Virais Reguladoras e Acessórias/farmacologia , Vírion/fisiologia , Gencitabina
8.
Future Microbiol ; 7(9): 1117-26, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22953710

RESUMO

Some intracellular/membranous factors exert intrinsic immunity against viral pathogens. Most recently, SAMHD1 has been shown to be one of these factors. SAMHD1 is a nucleus-localized protein, and mutations in the gene are associated with Aicardi-Goutières syndrome. As a triphosphohydrolase, it depletes the intracellular pool of dNTPs in myeloid cells, such as macrophages and dendritic cells, to a low level that establishes a precursor-deficient environment for the synthesis of lentiviral cDNA, thereby restricting viral replication in these host cells. However, some viruses evolve Vpx to recruit SAMHD1 onto the CRL4(DCAF1) E3 ubiquitin ligase in the cytoplasm for proteasome-dependent degradation, by which these viruses relieve SAMHD1-mediated restriction of primate lentivirus infection. In this review, we describe the latest knowledge of SAMHD1 biology.


Assuntos
Antivirais/farmacologia , Células Dendríticas/virologia , HIV-1/efeitos dos fármacos , Macrófagos/virologia , Proteínas Monoméricas de Ligação ao GTP/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Doenças Autoimunes do Sistema Nervoso/genética , Doenças Autoimunes do Sistema Nervoso/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , HIV-1/genética , HIV-1/fisiologia , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Proteínas Monoméricas de Ligação ao GTP/química , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/metabolismo , Proteína 1 com Domínio SAM e Domínio HD , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/metabolismo , Proteínas Virais Reguladoras e Acessórias/farmacologia
9.
PLoS One ; 6(11): e27660, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22110710

RESUMO

Among its many roles, the HIV-1 accessory protein Vpu performs a viroporin function and also antagonizes the host cell restriction factor tetherin through its transmembrane domain. BIT225 is a small molecule inhibitor that specifically targets the Vpu viroporin function, which, in macrophages, resulted in late stage inhibition of virus release and decreased infectivity of released virus, a phenotype similar to tetherin-mediated restriction. Here, we investigated whether BIT225 might mediate its antiviral function, at least in part, via inhibition of Vpu-mediated tetherin antagonism. Using T-cell lines inducible for tetherin expression, we found that BIT225 does not exert its antiviral function by inhibiting Vpu-mediated tetherin downmodulation from the cell surface, the main site of action of tetherin activity. In addition, results from a bioluminescence resonance energy transfer (BRET) assay showed that the Vpu-tetherin interaction was not affected by BIT225. Our data provide support for the concept that tetherin antagonism and viroporin function are separable on the Vpu transmembrane and that viroporin function might be cell-type dependent. Further, this work contributes to the characterization of BIT225 as an inhibitor that specifically targets the viroporin function of Vpu.


Assuntos
Fármacos Anti-HIV/farmacologia , Guanidina/farmacologia , Guanidinas/farmacologia , HIV-1/fisiologia , Proteínas do Vírus da Imunodeficiência Humana/antagonistas & inibidores , Proteínas do Vírus da Imunodeficiência Humana/farmacologia , Pirazóis/farmacologia , Proteínas Virais Reguladoras e Acessórias/antagonistas & inibidores , Proteínas Virais Reguladoras e Acessórias/farmacologia , Antígenos CD/metabolismo , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas Ligadas por GPI/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , HIV-1/efeitos dos fármacos , HIV-1/metabolismo , Proteínas do Vírus da Imunodeficiência Humana/química , Humanos , Estrutura Terciária de Proteína , Proteínas Virais Reguladoras e Acessórias/química , Liberação de Vírus/efeitos dos fármacos
10.
Blood ; 117(24): 6600-7, 2011 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-21521785

RESUMO

HIV-1 viral protein U (Vpu) is involved in ubiquitination and degradation of BM stromal cell Ag 2 and surface receptor CD4 through their recruitment to SCF(ß-TrcP) (Skp1/Cul1/F-box) ubiquitin ligase (SCF) complex. Here, we show that specific interaction of wild-type Vpu protein with SCF complex leads to inhibition of ubiquitination and proteasomal degradation of p53 protein in a ß-TrcP-dependent manner. Successful interaction of SCF(ß-TrcP) complex with ß-TrcP binding motif (DS(52)GNES(56)) present in Vpu is essential because mutant Vpu possessing specific alanine substitutions (DA(52)GNEA(56)) in the ß-TrcP binding motif not only failed to stabilize p53 protein but was also unable to inhibit ubiquitination of p53 protein. Furthermore, Vpu competes efficiently with the interaction of p53 protein with the ß-TrcP subunit of the SCF complex and inhibits subsequent ubiquitination of p53 proteins in a dose-dependent manner. We also observed potent apoptotic activity in a p53 null cell line (H-1299) that was cotransfected with p53 and Vpu-expressing plasmids. Furthermore, MOLT-3 (human T-lymphoblast) cells when infected with vesicular stomatitis virus glycoprotein-pseudotypic HIV-1 possessing wild-type vpu gene exhibited maximum activation of p53/Bax proteins and p53-mediated cell death. These findings establish a novel function of Vpu in modulating the stability of p53 protein that correlates positively with apoptosis during late stages of HIV-1 infection.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas do Vírus da Imunodeficiência Humana/farmacologia , Linfócitos T/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/fisiologia , Ubiquitinação/efeitos dos fármacos , Proteínas Virais Reguladoras e Acessórias/farmacologia , Proteínas Contendo Repetições de beta-Transducina/antagonistas & inibidores , Apoptose/fisiologia , Células Cultivadas , Infecções por HIV/metabolismo , Infecções por HIV/patologia , HIV-1/genética , HIV-1/metabolismo , HIV-1/fisiologia , Proteínas do Vírus da Imunodeficiência Humana/genética , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Proteínas do Vírus da Imunodeficiência Humana/fisiologia , Humanos , Células K562 , Ligação Proteica/genética , Domínios e Motivos de Interação entre Proteínas/genética , Domínios e Motivos de Interação entre Proteínas/fisiologia , Estabilidade Proteica/efeitos dos fármacos , Linfócitos T/metabolismo , Linfócitos T/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/metabolismo , Proteínas Virais Reguladoras e Acessórias/fisiologia , Proteínas Contendo Repetições de beta-Transducina/química , Proteínas Contendo Repetições de beta-Transducina/genética , Proteínas Contendo Repetições de beta-Transducina/metabolismo
11.
J Mol Med (Berl) ; 87(9): 899-911, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19579018

RESUMO

Novel treatment modalities, including gene therapy, are needed for patients with advanced melanoma. The E gene from the phage varphiX174 encodes a 91-aa protein which lyses Escherichia coli by formation of a transmembrane tunnel structure. To evaluate whether this E gene has a cytotoxic impact on melanoma cells in vitro and in vivo, and could therefore be used as a new therapeutic strategy for this tumor type, we selected the B16-F10 murine melanoma cell line as a model. We used a nonviral gene delivery approach (pcDNA3.1/E plasmid) to study the inhibition of melanoma cells' proliferation in vitro and direct intratumoral injection of pcDNA3.1/E complexed with jetPEI to deliver E cDNA to rapidly growing murine melanomas, and found that the E gene has both a strong antiproliferative effect in B16-F10 cells in vitro and induces an efficient decrease in melanoma tumor volume in vivo (90% in 15 days). Interestingly, the GFP-E fusion protein expressed in melanoma cells was located in the mitochondria. In vitro and in vivo analysis demonstrated significant functional and morphological mitochondrial alterations accompanied by a significant increase of cytochrome c and active caspase-3 and -9 in transfected cells, which suggests that tumoral cell death is mediated by the mitochondrial apoptotic pathway. These results show that E gene expression in melanoma cells has an extraordinary antitumor effect, which means it may be a new candidate for an effective strategy for melanoma treatment.


Assuntos
Antineoplásicos/farmacologia , Proteínas Repressoras/farmacologia , Proteínas Virais Reguladoras e Acessórias/farmacologia , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , DNA Viral/administração & dosagem , Melanoma Experimental , Camundongos , Mitocôndrias/efeitos dos fármacos , Proteínas Mitocondriais/efeitos dos fármacos , Proteínas Repressoras/uso terapêutico , Proteínas Virais Reguladoras e Acessórias/uso terapêutico
12.
Virol J ; 5: 60, 2008 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-18489774

RESUMO

BACKGROUND: HIV-1 mediated perturbation of the plasma membrane can produce an alteration in the transmembrane gradients of cations and other small molecules leading to cell death. Several HIV-1 proteins have been shown to perturb membrane permeability and ion transport. Xenopus laevis oocytes have few functional endogenous ion channels, and have proven useful as a system to examine direct effects of exogenously added proteins on ion transport. RESULTS: HIV-1 Nef induces alterations in the intracellular potassium concentration in CD4+ T-lymphoblastoid cells, but not intracellular pH. Two electrode voltage-clamp recording was used to determine that Nef did not form ion channel-like pores in Xenopus oocytes. CONCLUSION: These results suggest that HIV-1 Nef regulates intracellular ion concentrations indirectly, and may interact with membrane proteins such as ion channels to modify their electrical properties.


Assuntos
Permeabilidade da Membrana Celular/efeitos dos fármacos , Líquido Intracelular/metabolismo , Potássio/metabolismo , Proteínas Virais Reguladoras e Acessórias/farmacologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/farmacologia , Animais , Linhagem Celular Tumoral , Humanos , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Líquido Intracelular/efeitos dos fármacos , Transporte de Íons/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Proteínas Recombinantes/farmacologia , Xenopus laevis
13.
BMC Biotechnol ; 8: 43, 2008 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-18439318

RESUMO

BACKGROUND: Thermostable enzymes from thermophiles have attracted extensive studies. In this investigation, a nuclease-encoding gene (designated as GBSV1-NSN) was obtained from a thermophilic bacteriophage GBSV1 for the first time. RESULTS: After recombinant expression in Escherichia coli, the purified GBSV1-NSN exhibited non-specific nuclease activity, being able to degrade various nucleic acids, including RNA, single-stranded DNA and double-stranded DNA that was circular or linear. Based on sequence analysis, the nuclease shared no homology with any known nucleases, suggesting that it was a novel nuclease. The characterization of the recombinant GBSV1-NSN showed that its optimal temperature and pH were 60 degrees C and 7.5, respectively. The results indicated that the enzymatic activity was inhibited by enzyme inhibitors or detergents, such as ethylene diamine tetraacetic acid, citrate, dithiothreitol, beta-mercaptoethanol, guanidine hydrochloride, urea and SDS. In contrast, the nuclease activity was enhanced by TritonX-100, Tween-20 or chaps to approximately 124.5% - 141.6%. The Km of GBSV1-NSN nuclease was 231, 61 and 92 microM, while its kcat was 1278, 241 and 300 s-1 for the cleavage of dsDNA, ssDNA and RNA, respectively. CONCLUSION: Our study, therefore, presented a novel thermostable non-specific nuclease from thermophilic bacteriophage and its overexpression and purification for scientific research and applications.


Assuntos
Caudovirales/enzimologia , Endonucleases/farmacologia , Ácidos Nucleicos/metabolismo , Proteínas Virais Reguladoras e Acessórias/farmacologia , Caudovirales/genética , Caudovirales/isolamento & purificação , Clonagem Molecular , Endonucleases/antagonistas & inibidores , Endonucleases/genética , Endonucleases/metabolismo , Ativadores de Enzimas/farmacologia , Inibidores Enzimáticos/farmacologia , Estabilidade Enzimática , Escherichia coli/genética , Proteínas de Escherichia coli/biossíntese , Proteínas de Escherichia coli/isolamento & purificação , Exonucleases/antagonistas & inibidores , Exonucleases/genética , Exonucleases/metabolismo , Exonucleases/farmacologia , Expressão Gênica , Cinética , Proteínas Recombinantes de Fusão , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Temperatura , Proteínas Virais Reguladoras e Acessórias/genética
14.
J Virol ; 81(24): 13852-64, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17928336

RESUMO

Nef is a multifunctional accessory protein of primate lentiviruses. Recently, it has been shown that the ability of Nef to downmodulate CD4, CD28, and class I major histocompatibility complex is highly conserved between most or all primate lentiviruses, whereas Nef-mediated downregulation of T-cell receptor-CD3 was lost in the lineage that gave rise to human immunodeficiency virus type 1 (HIV-1). Whether or not other Nef activities are preserved between different groups of primate lentiviruses remained to be determined. Here, we show that nef genes from a large variety of HIVs and simian immunodeficiency viruses (SIVs) enhance virion infectivity and stimulate viral replication in human cells and/or in ex vivo infected human lymphoid tissue (HLT). Notably, nef alleles from unpassaged SIVcpz and SIVsmm enhanced viral infectivity, replication, and cytopathicity in cell culture and in ex vivo infected HLT as efficiently as those from HIV-1 and HIV-2, their human counterparts. Furthermore, nef genes from several highly divergent SIVs that have not been found in humans were also highly active in human cells and/or tissues. Thus, most primate lentiviral Nefs enhance virion infectivity and stimulate viral replication. Moreover, our data show that SIVcpz and SIVsmm Nefs do not require adaptive changes to perform these functions in human cells or tissues and support the idea that nef alleles from other primate lentiviruses would also be capable of promoting efficient virus spread in humans.


Assuntos
HIV-1/patogenicidade , Vírus da Imunodeficiência Símia/patogenicidade , Proteínas Virais Reguladoras e Acessórias/metabolismo , Vírion/patogenicidade , Replicação Viral/efeitos dos fármacos , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Animais , Linhagem Celular , HIV-1/classificação , HIV-1/genética , Células HeLa , Humanos , Técnicas de Cultura de Órgãos , Tonsila Palatina/virologia , Vírus da Imunodeficiência Símia/classificação , Vírus da Imunodeficiência Símia/genética , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/farmacologia , Replicação Viral/ética , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética
15.
Ann N Y Acad Sci ; 1056: 279-92, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16387695

RESUMO

Evidence is presented to suggest that HIV-1 accessory protein Nef could be involved in AIDS pathogenesis. When present in extracellular medium, Nef causes the death of a wide variety of cells in vitro and may therefore be responsible for the depletion of bystander cells in lymphoid tissues during HIV infection. When present inside the cell, Nef could prevent the death of infected cells and thereby contribute to increased viral load. Intracellular Nef does this by preventing apoptosis of infected cells by either inhibiting proteins involved in apoptosis or preventing the infected cells from being recognized by CTLs. Neutralization of extracellular Nef could prevent the death of uninfected immune cells and thereby the destruction of the immune system. Neutralization of intracellular Nef could hasten the death of infected cells and help reduce the viral load. Nef is therefore a very important molecular target for developing therapeutics that slow progression to AIDS. The N-terminal region of Nef and the naturally occurring bee venom mellitin have very similar primary and tertiary structures, and they both act by destroying membranes. Chemical analogs of a mellitin inhibitor prevent Nef-mediated cell death and inhibit the interaction of Nef with cellular proteins involved in apoptosis. Naturally occurring bee propolis also contains substances that prevent Nef-mediated cell lysis and increases proliferation of CD4 cells in HIV-infected cultures. These chemical compounds and natural products are water soluble and nontoxic and are therefore potentially very useful candidate drugs.


Assuntos
Vacinas contra a AIDS , Síndrome da Imunodeficiência Adquirida/imunologia , Produtos do Gene nef/farmacologia , Síndrome da Imunodeficiência Adquirida/fisiopatologia , Sequência de Aminoácidos , Produtos do Gene nef/química , Produtos do Gene nef/imunologia , Humanos , Meliteno/química , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Proteínas Virais/metabolismo , Proteínas Virais Reguladoras e Acessórias/imunologia , Proteínas Virais Reguladoras e Acessórias/farmacologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana
16.
Genes Dev ; 11(17): 2204-13, 1997 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9303536

RESUMO

One of the classical positive regulators of gene expression is bacteriophage lambda N protein. N regulates the transcription of early phage genes by participating in the formation of a highly processive, terminator-resistant transcription complex and thereby stimulates the expression of genes lying downstream of transcriptional terminators. Also included in this antiterminating transcription complex are an RNA site (NUT) and host proteins (Nus). Here we demonstrate that N has an additional, hitherto unknown regulatory role, as a repressor of the translation of its own gene. N-dependent repression does not occur when NUT is deleted, demonstrating that N-mediated antitermination and translational repression both require the same cis-acting site in the RNA. In addition, we have identified one nut and several host mutations that eliminate antitermination and not translational repression, suggesting the independence of these two N-mediated mechanisms. Finally, the position of nutL with respect to the gene whose expression is repressed is important.


Assuntos
Biossíntese de Proteínas/efeitos dos fármacos , Fatores de Transcrição/farmacologia , Proteínas Virais Reguladoras e Acessórias/farmacologia , Bacteriófago lambda/efeitos dos fármacos , Bacteriófago lambda/genética , Bacteriófago lambda/metabolismo , Sequência de Bases , Mapeamento Cromossômico , Primers do DNA/genética , Regulação Viral da Expressão Gênica , Genes Reporter , Genes Virais , Modelos Genéticos , Dados de Sequência Molecular , Mutação , Conformação de Ácido Nucleico , Reação em Cadeia da Polimerase , RNA Viral/química , RNA Viral/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/metabolismo
17.
EMBO J ; 15(22): 6155-65, 1996 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-8947037

RESUMO

The vpr genes of human and simian immunodeficiency viruses (HIV/SIV) encode proteins which are packaged in the virus particle. HIV-1 Vpr has been shown to mediate the nuclear import of viral reverse transcription complexes in non-dividing target cells (e.g. terminally differentiated macrophages), and to alter the cell cycle and proliferation status of the infected host cell. Members of the HIV-2/SIV(SM) group encode, in addition to Vpr, a related protein called Vpx. Because these two proteins share considerable sequence similarity, it has been assumed that they also exhibit similar functions. Here, we report that the functions of Vpr and Vpx are distinct and non-redundant, although both proteins are components of the HIV-2/SIV(SM) virion and reverse transcription complex. Characterizing SIV(SM) proviruses defective in one or both genes, we found that Vpx is both necessary and sufficient for the nuclear import of the viral reverse transcription complex. In contrast, Vpr, but not Vpx, inhibited the progression of infected host cells from the G2 to the M phase of the cell cycle. Thus, two independent functions of the HIV-1 Vpr protein are encoded by separate genes in HIV-2/SIV(SM). This segregation is consistent with the conservation of these genes in HIV-2/SIV(SM) evolution, and underscores the importance of both nuclear transport and cell cycle arrest functions in primate lentivirus biology.


Assuntos
Produtos do Gene vpr/metabolismo , Genes Virais/genética , HIV-2/metabolismo , Animais , Western Blotting , Ciclo Celular/genética , Divisão Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Células Cultivadas , DNA Viral/metabolismo , Produtos do Gene gag/metabolismo , Produtos do Gene vpr/farmacologia , HIV-2/genética , Haplorrinos , Macrófagos/metabolismo , Macrófagos/virologia , Mutagênese Sítio-Dirigida/genética , DNA Polimerase Dirigida por RNA/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Proteínas Virais Reguladoras e Acessórias/farmacologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana
18.
Cell ; 78(2): 317-24, 1994 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-8044843

RESUMO

A prolonged pause in transcription elongation at positions +16 and +17 of the phage lambda late gene operon has an important role in the modification of RNA polymerase by the lambda gene Q transcription antiterminator. Mutations included in the transcription bubble of the paused transcription complex, particularly at +2 and +6, abolish pausing and the ability of Q protein to modify RNA polymerase. By transcribing heteroduplex templates made in vitro, we show that the sites identified by these mutations act through the nontranscribed strand of DNA. This result suggests unexpected regulatory functions of the nontranscribed DNA strand in transcription.


Assuntos
Bacteriófago lambda/genética , DNA Antissenso/genética , DNA Viral/genética , Transcrição Gênica/fisiologia , Sequência de Bases , DNA Antissenso/fisiologia , DNA Viral/fisiologia , RNA Polimerases Dirigidas por DNA/metabolismo , Regulação Viral da Expressão Gênica/fisiologia , Genes Virais/fisiologia , Dados de Sequência Molecular , Mutação Puntual/fisiologia , RNA Mensageiro/biossíntese , Transcrição Gênica/efeitos dos fármacos , Proteínas Virais Reguladoras e Acessórias/farmacologia
19.
J Virol ; 67(12): 7238-45, 1993 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8230446

RESUMO

Two functions have been attributed to the product of the human immunodeficiency virus type 1 vpu open reading frame: it increases virion release from infected cells and induces rapid degradation of CD4 shortly after its synthesis. In the absence of Vpu, newly synthesized gp160 and CD4 associate in the endoplasmic reticulum (ER), forming a complex whose further maturation is blocked and which is eventually degraded. In studies using NL4-3-based expression vectors, it has been previously shown that Vpu induces the release of gp160 from the complex that it forms with CD4 in the ER. This release, which appears to be due to the rapid degradation of CD4 induced by Vpu, allows gp160 to transit to the Golgi, where it matures further. We investigated which regions of CD4 are important for its susceptibility to Vpu-induced degradation by transfecting HeLa cells with isogenic vpu-positive and vpu-negative proviruses and vectors expressing various truncated or mutated CD4 molecules. The results suggested that the cytoplasmic domain of CD4 contains a determinant lying within amino acids 418 to 425 that is critical for susceptibility to Vpu-induced degradation. Neither the phosphorylation sites in the cytoplasmic domain nor the Lck interaction region was required for the effect. Vpu-induced degradation was specific for CD4, since CD8, even when retained in the ER, was not degraded. In addition, under conditions of high-level Vpu expression, CD4 degradation could be observed in the absence of gp160 or other means of retaining CD4 in the ER.


Assuntos
Antígenos CD4/metabolismo , HIV-1/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Sequência de Aminoácidos , Antígenos CD4/efeitos dos fármacos , Códon , Análise Mutacional de DNA , Produtos do Gene env/genética , Produtos do Gene env/metabolismo , Proteína gp160 do Envelope de HIV , HIV-1/genética , Células HeLa , Proteínas do Vírus da Imunodeficiência Humana , Humanos , Dados de Sequência Molecular , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Especificidade por Substrato , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/farmacologia
20.
J Virol ; 67(12): 6956-64, 1993 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8230418

RESUMO

The RAW264 murine macrophage cell line was used as a model to examine the role of the tat and nef gene products in the transcription regulation of the human immunodeficiency virus type 1 (HIV-1) long terminal repeat (LTR) in macrophages. Contrary to claims that the activity of the HIV-1 LTR responds poorly in rodent cells to trans activation by the viral tat gene product, cotransfection of RAW264 cells with a tat expression plasmid in transient transfection assays caused a > 20-fold increase in reporter gene expression that was inhibited by mutations in the TAR region. RAW264 cells stably transfected with the tat plasmid displayed similarly elevated HIV-1 LTR-driven reporter gene activity. By contrast to previous reports indicating a negative role for nef in HIV transcription, cotransfection of RAW264 cells with a nef expression plasmid trans activated the HIV-1 LTR driving either a chloramphenicol acetyltransferase or a luciferase reporter gene. The action of nef was specific to the LTR, as expression of nef had no effect on the activity of the simian virus 40, c-fms, urokinase plasminogen activator, or type 5 acid phosphatase promoter. trans-activating activity was also manifested by a frameshift mutant expressing only the first 35 amino acids of the protein. The effects of nef were multiplicative with those of tat gene product and occurred even in the presence of bacterial lipopolysaccharide, which itself activated LTR-directed transcription. Examination of the effects of selected mutations in the LTR revealed that neither the kappa B sites in the direct repeat enhancer nor the TAR region was required as a cis-acting element in nef action. The action of nef was not species restricted; it was able to trans activate in the human monocyte-like cell line Mono Mac 6. The presence of a nef expression cassette in a neomycin phosphotransferase gene expression plasmid greatly reduced the number of G418-resistant colonies generated in stable transfection of RAW264 cells, and many of the colonies that were formed exhibited very slow growth. The frameshift mutant was also active in reducing colony generation. Given the absence of any effect of the frameshift mutation on nef function, its actions on macrophage growth and HIV transcription are discussed in terms of the role of the N-terminal 30 amino acids and of stable secondary structures in the mRNA.


Assuntos
Repetição Terminal Longa de HIV/genética , HIV-1/genética , Macrófagos/microbiologia , Transcrição Gênica/efeitos dos fármacos , Proteínas Virais Reguladoras e Acessórias/farmacologia , Animais , Linhagem Celular , Mutação da Fase de Leitura , Regulação Viral da Expressão Gênica , Produtos do Gene nef/farmacologia , Produtos do Gene tat/farmacologia , Genes Reporter , HIV-1/crescimento & desenvolvimento , Haplorrinos , Humanos , Lipopolissacarídeos/farmacologia , Camundongos , Conformação de Ácido Nucleico , RNA Mensageiro/genética , Sequências Reguladoras de Ácido Nucleico/genética , Vírus 40 dos Símios/genética , Especificidade da Espécie , Transfecção , Produtos do Gene nef do Vírus da Imunodeficiência Humana , Produtos do Gene tat do Vírus da Imunodeficiência Humana
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA