Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Virol J ; 21(1): 26, 2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-38263162

RESUMO

BACKGROUND: Infection by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and influenza virus is still a major worldwide health concern. Plants are a good source of bioactive compounds to be used as preventive measures for both inhibiting the virus binding and enhancing mucosal innate immunity. Curcumin has been shown to possess antiviral activity and modulate innate immunity. Therefore, the purpose of this study was to develop an oro-nasal film spray containing curcumin and determine its antiviral activity against SARS-CoV-2 and influenza virus infection, as well as its effects on mucosal innate immunity and inflammatory cytokines in vitro. METHODS: The antiviral activity of the film spray against SARS-CoV-2, influenza A/H1N1, A/H3N2, and influenza B was assessed in vitro by plaque reduction assay. Cytotoxicity of the film spray to oral keratinocytes and nasal epithelial cells was assessed by MTT assay, and cytotoxicity to Vero and MDCK cells was assessed by an MTS-based cytotoxicity assay. Oral and nasal innate immune markers in response to the film spray were determined by ELISA and by a commercial Milliplex Map Kit, respectively. RESULTS: Our data show that the film spray containing curcumin can inhibit both SARS-CoV-2 and influenza virus infections while maintaining cell viability. Results obtained among 4 viruses revealed that curcumin film spray demonstrated the highest inhibitory activity against SARS-CoV-2 with the lowest EC50 of 3.15 µg/ml and the highest SI value of 4.62, followed by influenza B (EC50 = 6.32 µg/ml, SI = 2.04), influenza A/H1N1 (EC50 = 7.24 µg/ml, SI = 1.78), and influenza A/H3N2 (EC50 > 12.5 µg/ml, SI < 1.03), respectively. Antimicrobial peptides LL-37 and HD-5, IL-6 and TNF-α produced by oral keratinocytes were significantly induced by the film spray, while hBD2 was significantly reduced. CONCLUSION: Film spray containing curcumin possesses multiple actions against SARS-CoV-2 infection by inhibiting ACE-2 binding in target cells and enhancing mucosal innate immunity. The film spray can also inhibit influenza virus infection. Therefore, the curcumin film spray may be effective in preventing the viral infection of both SARS-CoV-2 and influenza.


Assuntos
COVID-19 , Curcumina , Vírus da Influenza A Subtipo H1N1 , Influenza Humana , Infecções por Orthomyxoviridae , Animais , Cães , Humanos , SARS-CoV-2 , Imunidade nas Mucosas , Vírus da Influenza A Subtipo H3N2 , Células Madin Darby de Rim Canino , Antivirais
2.
Pharmaceutics ; 14(12)2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36559173

RESUMO

Favipiravir (FVR) is a repurposed antiviral drug for treating mild to moderate cases of the novel coronavirus disease 2019 (COVID-19). However, its poor solubility and permeability limit its clinical efficacy. To overcome its physicochemical and pharmacokinetic limitations, we statistically designed a mucoadhesive chitosan-alginate nanoparticles (MCS-ALG-NPs) as a new carrier for FVR using response surface methodology, which provided suitable characteristics for transmucosal delivery. The use of mucoadhesive polymers for intranasal administration promotes the residence time and contact of FVR in the mucus membrane. The optimized FVR-MCS-ALG-NPs demonstrated superior mucoadhesion, higher permeation and deposition in the nasal mucosa, and a significant increase in the inhibition of viral replication over 35-fold compared with free FVR. The overall results suggest that MCS-ALG-NPs could be used as an effective mucoadhesive carrier to enhance the activity of FVR against COVID-19.

3.
Pharmaceuticals (Basel) ; 15(5)2022 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-35631446

RESUMO

Since the pandemic of severe acute respiratory syndrome coronavirus (SARS-CoV-2) in December 2019, the infection cases have quickly increased by more than 511 million people. The long epidemic outbreak over 28 months has affected health and economies worldwide. An alternative medicine appears to be one choice to alleviate symptoms and reduce mortality during drug shortages. Dendrobium extract is one of the traditional medicines used for COVID-19 infection. Several compounds in Dendrobium sp. had been reported to exert pharmacological activities to treat common COVID-19-related symptoms. Herein, in silico screening of 83 compounds from Dendrobium sp. by using the SARS-CoV-2 spike protein receptor-binding domain (RBD) as a drug target was performed in searching for a new lead compound against SARS-CoV-2 infection. Four hit compounds showing good binding affinity were evaluated for antiviral infection activity. The new lead compound DB36, 5-methoxy-7-hydroxy-9,10-dihydro-1,4-phenanthrenequinone, was identified with the IC50 value of 6.87 ± 3.07 µM. The binding mode revealed that DB36 bound with the spike protein at the host receptor, angiotensin-converting enzyme 2 (ACE2) binding motif, resulted in antiviral activity. This study substantiated the use of Dendrobium extract for the treatment of SARS-CoV-2 infection and has identified new potential chemical scaffolds for further drug development of SARS-CoV-2 entry inhibitors.

4.
Comput Struct Biotechnol J ; 20: 882-890, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35136534

RESUMO

The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 2019 has posed a serious threat to global health and the economy for over two years, prompting the need for development of antiviral inhibitors. Due to its vital role in viral replication, RNA-dependent RNA polymerase (RdRp) is a promising therapeutic target. Herein, we analyzed amino acid sequence conservation of RdRp across coronaviruses. The conserved amino acids at the catalytic binding site served as the ligand-contacting residues for in silico screening to elucidate possible resistant mutation. Molecular docking was employed to screen inhibitors of SARS-CoV-2 from the ZINC ChemDiv database. The top-ranked compounds selected from GOLD docking were further investigated for binding modes at the conserved residues of RdRp, and ten compounds were selected for experimental validation. Of which, three compounds exhibited promising antiviral activity. The most promising candidate showed a half-maximal effective concentration (EC50) of 5.04 µM. Molecular dynamics simulations, binding free-energy calculation and hydrogen bond analysis were performed to elucidate the critical interactions providing a foundation for developing lead compounds effective against SARS-CoV-2.

5.
Foods ; 10(12)2021 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-34945591

RESUMO

Coccinia grandis or ivy gourd is an edible plant. Its leaves and fruits are used as vegetable in many countries. Many works on antidiabetic activity of a crude extract of C. grandis, i.e., in vitro, in vivo, and clinical trials studies, have been reported. Profiles of the antidiabetic compounds were previously proposed by using LC-MS or GC-MS. However, the compounds responsible for antidiabetic activity have rarely been isolated and characterized by analysis of 1D and 2D NMR data. In the present work, UHPLC-ESI-QTOF-MS/MS analysis and GNPS molecular networking were used to guide the isolation of α-glucosidase inhibitors from an extract of C. grandis leaves. Seven flavonoid glycosides including rutin (1), kaempferol 3-O-rutinoside (2) or nicotiflorin, kaempferol 3-O-robinobioside (3), quercetin 3-O-robinobioside (4), quercetin 3-O-ß-D-apiofuranosyl-(1→2)-[α-L-rhamnopyranosyl-(1→6)]-ß-D-glucopyranoside (5) or CTN-986, kaempferol 3-O-ß-D-api-furanosyl-(1→2)-[α-L-rhamnopyranosyl-(1→6)]-ß-D-glucopyranoside (6), and kaempferol 3-O-ß-D-apiofuranosyl-(1→2)-[α-L-rhamnopyranosyl-(1→6)]-ß-D-galactopyranoside (7) were isolated from C. grandis leaves. This is the first report of glycosides containing apiose sugar in the genus Coccinia. These glycosides exhibited remarkable α-glucosidase inhibitory activity, being 4.4-10.3 times more potent than acarbose. Moreover, they also displayed virucidal activity against influenza A virus H1N1, as revealed by the ASTM E1053-20 method.

6.
Viruses ; 13(11)2021 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-34835114

RESUMO

Human immunodeficiency virus type 1 (HIV-1) modulates the host cell cycle. The HIV-1 accessory protein Vpr arrests the cell cycle at the G2 phase in dividing cells, and the ability of Vpr to induce G2 arrest is well conserved among primate lentiviruses. Additionally, Vpr-mediated G2 arrest likely correlates with enhanced HIV-1 infection in monocyte-derived macrophages. Here, we screened small-interfering RNA to reveal candidates that suppress Vpr-induced G2 arrest and identified Huntingtin-interacting protein 1 (HIP1) required for efficient G2 arrest. Interestingly, HIP1 was not essential for Vpr-induced DNA double-strand breaks, which are required for activation of the DNA-damage checkpoint and G2 arrest. Furthermore, HIP1 knockdown suppressed HIV-1 infection in monocyte-derived macrophages. This study identifies HIP1 as a factor promoting Vpr-induced G2 arrest and HIV-1 infection in macrophages.


Assuntos
Proteínas de Ligação a DNA/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , RNA Interferente Pequeno , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia , Pontos de Checagem do Ciclo Celular , Células HEK293 , Células HeLa , Humanos , Macrófagos , Monócitos , Replicação Viral
7.
Biochem Biophys Res Commun ; 503(4): 2970-2976, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30126636

RESUMO

The interaction between viral protein Gag and cellular protein tumor susceptibility gene 101 (TSG101) is a crucial step in the HIV-1 replication cycle. This interaction initiates the viral assembly/budding via the cellular endosomal sorting complexes required for transport (ESCRT) pathway, making it a potential target for antiviral therapy. Here we developed a simple, robust, and reliable high-throughput screening (HTS) system based on enzyme-linked immunosorbent assay (ELISA) to identify compounds that inhibit HIV-1 replication by targeting Gag-TSG101 interaction. Through screening of the 9600-compound library using the established HTS system, several hit compounds, which inhibited Gag-TSG101 interaction, were identified. Subsequent assays revealed two hit compounds, HSM-9 and HSM-10, which have antiviral activity against CD4+ T cell-tropic NL4-3 and macrophage-tropic JR-CSF HIV-1 strains. These results suggest that our established HTS system is an indispensable tool for the identification of HIV-1 Gag-TSG101 interaction inhibitors.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , HIV-1 , Fatores de Transcrição/metabolismo , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Ligação Proteica/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
8.
Virology ; 507: 32-39, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28399435

RESUMO

An anti-influenza compound, DP2392-E10 based on inhibition of the nuclear export function of the viral nucleoprotein-nuclear export signal 3 (NP-NES3) domain was successfully identified by our previous high-throughput screening system. Here, we demonstrated that DP2392-E10 exerts its antiviral effect by inhibiting replication of a broad range of influenza A subtypes. In regard to the molecular mechanism, we revealed that DP2392-E10 inhibits nuclear export of both viral NP and nuclear export protein (NEP). More specifically, in vitro pull-down assays revealed that DP2392-E10 directly binds cellular CRM1, which mediates nuclear export of NP and NEP. In silico docking suggested that DP2392-E10 binds at a region close to the HEAT9 and HEAT10 domains of CRM1. Together, these results indicate that the CRM1-mediated nuclear export function of influenza virus represents a new potential target for antiviral drug development, and also provide a core structure for a novel class of inhibitors that target this function.


Assuntos
Vírus da Influenza A/metabolismo , Influenza Humana/metabolismo , Carioferinas/metabolismo , Nucleoproteínas/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Antivirais/farmacologia , Linhagem Celular , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Humanos , Vírus da Influenza A/genética , Influenza Humana/virologia , Carioferinas/antagonistas & inibidores , Carioferinas/genética , Nucleoproteínas/genética , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/genética
9.
PLoS One ; 11(9): e0163100, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27648839

RESUMO

HIV-1 budding requires interaction between Gag and cellular TSG101 to initiate viral particle assembly and release via the endosomal sorting complexes required for transport (ESCRT) pathway. However, some reports show that overexpression of TSG101 inhibits virus release by disruption of Gag targeting process. Since a HIV-1 accessory protein, Vpr binds to Gag p6 domain at the position close to the binding site for TSG101, whether Vpr implicates TSG101 overexpression effect has not been investigated. Here, we found that Vpr abrogates TSG101 overexpression effect to rescue viral production. Co-transfection of TSG101 and Gag with Vpr prevented TSG101-induced Gag accumulation in endosomes and lysosomes. In addition, Vpr rescued virus-like particle (VLP) production in a similar manner as a lysosomal inhibitor, Bafilomycin A1 indicating that Vpr inhibits TSG101-induced Gag downregulation via lysosomal pathway. Vpr and Gag interaction is required to counteract TSG101 overexpression effect since Vpr A30F mutant which is unable to interact with Gag and incorporate into virions, reduced ability to prevent Gag accumulation and to rescue VLP production. In addition, GST pull-down assays and Biacore analysis revealed that Vpr competed with TSG101 for Gag binding. These results indicate that Vpr overcomes the effects of TSG101 overexpression to support viral production by competing with TSG101 to bind Gag.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Produtos do Gene gag/metabolismo , HIV-1/fisiologia , Fatores de Transcrição/metabolismo , Liberação de Vírus/fisiologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo , Western Blotting , Proteínas de Ligação a DNA/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Endossomos/metabolismo , Transferência Ressonante de Energia de Fluorescência , Produtos do Gene gag/genética , Células HEK293 , HIV-1/genética , HIV-1/metabolismo , Células HeLa , Humanos , Lisossomos/metabolismo , Microscopia de Fluorescência/métodos , Mutação , Ligação Proteica , Fatores de Transcrição/genética , Vírion/genética , Vírion/metabolismo , Vírion/fisiologia , Montagem de Vírus/genética , Liberação de Vírus/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética
10.
Viruses ; 8(8)2016 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-27483302

RESUMO

Influenza remains a serious worldwide public health problem. After infection, viral genomic RNA is replicated in the nucleus and packed into viral ribonucleoprotein, which will then be exported to the cytoplasm via a cellular chromosome region maintenance 1 (CRM1)-dependent pathway for further assembly and budding. However, the nuclear export mechanism of influenza virus remains controversial. Here, we identify cellular nuclear transport factor 2 (NTF2)-like export protein 1 (NXT1) as a novel binding partner of nucleoprotein (NP) that stimulates NP-mediated nuclear export via the CRM1-dependent pathway. NXT1-knockdown cells exhibit decreased viral replication kinetics and nuclear accumulated viral RNA and NP. By contrast, NXT1 overexpression promotes nuclear export of NP in a CRM1-dependent manner. Pull-down assays suggest the formation of an NXT1, NP, and CRM1 complex, and demonstrate that NXT1 binds to the C-terminal region of NP. These findings reveal a distinct mechanism for nuclear export of the influenza virus and identify the NXT1/NP interaction as a potential target for antiviral drug development.


Assuntos
Transporte Ativo do Núcleo Celular , Interações Hospedeiro-Patógeno , Vírus da Influenza A Subtipo H1N1/fisiologia , Carioferinas/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Proteínas de Ligação a RNA/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas do Core Viral/metabolismo , Liberação de Vírus , Animais , Linhagem Celular , Cães , Humanos , Proteínas do Nucleocapsídeo , Montagem de Vírus
11.
PLoS One ; 10(12): e0145573, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26701275

RESUMO

The emergence of multidrug-resistant viruses compromises the efficacy of anti-human immunodeficiency virus type 1 (HIV-1) therapy and limits treatment options. Therefore, new targets that can be used to develop novel antiviral agents need to be identified. We previously identified a potential parent compound, hematoxylin, which suppresses the nuclear import of HIV-1 via the Vpr-importin α interaction and inhibits HIV-1 replication in a Vpr-dependent manner by blocking nuclear import of the pre-integration complex. However, it was unstable. Here, we synthesized a stable derivative of hematoxylin that bound specifically and stably to Vpr and inhibited HIV-1 replication in macrophages. Furthermore, like hematoxylin, the derivative inhibited nuclear import of Vpr in an in vitro nuclear import assay, but had no effect on Vpr-induced G2/M phase cell cycle arrest or caspase activity. Interestingly, this derivative bound strongly to amino acid residues 54-74 within the C-terminal α-helical domain (αH3) of Vpr. These residues are highly conserved among different HIV strains, indicating that this region is a potential target for drug-resistant HIV-1 infection. Thus, we succeeded in developing a stable hematoxylin derivative that bound directly to Vpr, suggesting that specific inhibitors of the interaction between cells and viral accessory proteins may provide a new strategy for the treatment of HIV-1 infection.


Assuntos
Fármacos Anti-HIV/síntese química , Infecções por HIV/prevenção & controle , HIV-1/efeitos dos fármacos , Hematoxilina/química , Macrófagos/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo , Fármacos Anti-HIV/farmacologia , Células Cultivadas , Infecções por HIV/virologia , Humanos , Macrófagos/virologia
12.
J Immunol ; 195(9): 4341-50, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26416279

RESUMO

Fibrocytes (fibroblastic leukocytes) are recently identified as unique hematopoietic cells with features of both macrophages and fibroblasts. Fibrocytes are known to contribute to the remodeling or fibrosis of various injured tissues. However, their role in viral infection is not fully understood. In this study, we show that differentiated fibrocytes are phenotypically distinguishable from macrophages but can be infected with HIV-1. Importantly, fibrocytes exhibited persistently infected cell-like phenotypes, the degree of which was more apparent than macrophages. The infected fibrocytes produced replication-competent HIV-1, but expressed HIV-1 mRNA at low levels and strongly resisted HIV-1-induced cell death, which enabled them to support an extremely long-term HIV-1 production at low but steady levels. More importantly, our results suggested that fibrocytes were susceptible to HIV-1 regardless of their differentiation state, in contrast to the fact that monocytes become susceptible to HIV-1 after the differentiation into macrophages. Our findings indicate that fibrocytes are the previously unreported HIV-1 host cells, and they suggest the importance of considering fibrocytes as one of the long-lived persistently infected cells for curing HIV-1.


Assuntos
Fibroblastos/virologia , HIV-1/fisiologia , Leucócitos/virologia , Macrófagos/virologia , Forma Celular/genética , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação Viral da Expressão Gênica , Infecções por HIV/sangue , HIV-1/genética , Interações Hospedeiro-Patógeno/genética , Humanos , Leucócitos/citologia , Leucócitos/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Microscopia Confocal , Monócitos/citologia , Monócitos/metabolismo , Monócitos/virologia , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Transcriptoma , Replicação Viral/genética
13.
PLoS One ; 9(8): e105081, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25119991

RESUMO

The assembly of influenza virus progeny virions requires machinery that exports viral genomic ribonucleoproteins from the cell nucleus. Currently, seven nuclear export signal (NES) consensus sequences have been identified in different viral proteins, including NS1, NS2, M1, and NP. The present study examined the roles of viral NES consensus sequences and their significance in terms of viral replication and nuclear export. Mutation of the NP-NES3 consensus sequence resulted in a failure to rescue viruses using a reverse genetics approach, whereas mutation of the NS2-NES1 and NS2-NES2 sequences led to a strong reduction in viral replication kinetics compared with the wild-type sequence. While the viral replication kinetics for other NES mutant viruses were also lower than those of the wild-type, the difference was not so marked. Immunofluorescence analysis after transient expression of NP-NES3, NS2-NES1, or NS2-NES2 proteins in host cells showed that they accumulated in the cell nucleus. These results suggest that the NP-NES3 consensus sequence is mostly required for viral replication. Therefore, each of the hydrophobic (Φ) residues within this NES consensus sequence (Φ1, Φ2, Φ3, or Φ4) was mutated, and its viral replication and nuclear export function were analyzed. No viruses harboring NP-NES3 Φ2 or Φ3 mutants could be rescued. Consistent with this, the NP-NES3 Φ2 and Φ3 mutants showed reduced binding affinity with CRM1 in a pull-down assay, and both accumulated in the cell nucleus. Indeed, a nuclear export assay revealed that these mutant proteins showed lower nuclear export activity than the wild-type protein. Moreover, the Φ2 and Φ3 residues (along with other Φ residues) within the NP-NES3 consensus were highly conserved among different influenza A viruses, including human, avian, and swine. Taken together, these results suggest that the Φ2 and Φ3 residues within the NP-NES3 protein are important for its nuclear export function during viral replication.


Assuntos
Vírus da Influenza A/fisiologia , Sinais de Exportação Nuclear , Nucleoproteínas/análise , Infecções por Orthomyxoviridae/virologia , Proteínas Virais/análise , Replicação Viral , Transporte Ativo do Núcleo Celular , Animais , Humanos , Vírus da Influenza A/genética , Influenza Humana/virologia , Mutação , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
14.
Biochem Biophys Res Commun ; 450(1): 49-54, 2014 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-24858693

RESUMO

Neuraminidase inhibitors are the only currently available influenza treatment, although resistant viruses to these drugs have already been reported. Thus, new antiviral drugs with novel mechanisms of action are urgently required. In this study, we identified a novel antiviral compound, WV970, through cell-based screening of a 50,000 compound library and subsequent lead optimization. This compound exhibited potent antiviral activity with nanomolar IC50 values against both influenza A and B viruses but not non-influenza RNA viruses. Time-of-addition and indirect immunofluorescence assays indicated that WV970 acted at an early stage of the influenza life cycle, but likely after nuclear entry of viral ribonucleoprotein (vRNP). Further analyses of viral RNA expression and viral polymerase activity indicated that WV970 inhibited vRNP-mediated viral genome replication and transcription. Finally, structure-based virtual screening and comprehensive human kinome screening were used to demonstrate that WV970 acts as a multiple kinase inhibitor, many of which are associated with influenza virus replication. Collectively, these results strongly suggest that WV970 is a promising anti-influenza drug candidate and that several kinases associated with viral replication are promising drug targets.


Assuntos
Inibidores da Síntese de Ácido Nucleico , Orthomyxoviridae/efeitos dos fármacos , Orthomyxoviridae/enzimologia , Fosfotransferases/antagonistas & inibidores , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Replicação Viral/fisiologia , Antivirais , Relação Dose-Resposta a Droga , Humanos , Concentração Inibidora 50 , Fosfotransferases/metabolismo , Replicação Viral/efeitos dos fármacos
15.
Microbes Infect ; 15(4): 280-90, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23376165

RESUMO

IFN-inducible IFITM proteins (IFITM1, 2, and 3) inhibit the replication of various viruses including HIV-1 through poorly understood mechanisms. Here, we further analyzed characteristics of these newly identified HIV-1 restriction factors. Firstly, in contrast to other anti-HIV-1 proteins, such as tetherin and APOBEC3G, IFITMs were resistant to a down-regulation of surface expression or degradation by HIV-1 proteins. Secondly, the enforced expression of IFITMs reduced the production of HIV-1 viruses from cells transfected with proviral plasmids containing whole viral sequences. Although their inhibitory activities were modest when compared to that of tetherin, IFITMs, but not tetherin, directly reduced the expression of HIV-1 proteins including Gag, Vif and Nef. Of importance, however, IFITMs had no inhibitory effect when these viral proteins were expressed by codon-optimized cDNAs that bypassed the viral-specific expression machinery. Indeed, our results supported the idea that IFITMs interfere with viral protein expression mediated by double-stranded viral RNAs, such as RRE and TAR. Finally, the S-palmitoylation of IFITMs, which is crucial for their anti-influenza virus activity, was not required for their anti-HIV-1 activity, indicating that IFITMs restrict these viruses at different steps. These characteristics lead to a better understanding of the mechanism by which IFITMs restrict HIV-1 and other viruses.


Assuntos
Antígenos de Diferenciação/imunologia , HIV-1/imunologia , HIV-1/fisiologia , Proteínas de Membrana/imunologia , Proteínas de Ligação a RNA/imunologia , Replicação Viral , Linhagem Celular , Humanos , Produtos do Gene gag do Vírus da Imunodeficiência Humana/antagonistas & inibidores , Produtos do Gene nef do Vírus da Imunodeficiência Humana/antagonistas & inibidores , Produtos do Gene vif do Vírus da Imunodeficiência Humana/antagonistas & inibidores
16.
J Immunol ; 188(8): 3620-7, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22407921

RESUMO

HIV-1 proteins, including Tat, gp120, and Nef, activate macrophages (MΦ), which is consistent with the fact that HIV-1 infection is characterized by sustained immune activation. Meanwhile, MΦ are functionally classified into two types: proinflammatory M1-MΦ and anti-inflammatory M2-MΦ. We show that HIV-1 proteins, particularly Nef, preferentially activate M2-MΦ. Extracellular Tat, gp120, and Nef activated MAPK and NF-κB pathways in human peripheral blood monocyte-derived MΦ. However, the activation was marked in M-CSF-derived M2-MΦ but not GM-CSF-derived M1-MΦ. Nef was the most potent activator, and its signaling activation was comparable to that by TNF-α. Indeed, Nef was internalized more rapidly by M2-MΦ than by M1-MΦ. The myristoylation and proline-rich motif of Nef were responsible for the observed signaling activation. Consistent with the activation of MAPK/NF-κB pathways, Nef stimulated the production of a number of proinflammatory cytokines/chemokines by M2-MΦ. However, Nef reduced the expression of CD163 and phagocytosis, the characteristic markers of M2-MΦ, indicating that Nef drives an M2-like to M1-like phenotypic shift. Because the differentiation of most tissue MΦ depends on M-CSF and its receptor, which is the essential axis for the anti-inflammatory M2-MΦ phenotype, the current study reveals an efficient mechanism by which HIV-1 proteins, such as Nef, induce the proinflammatory MΦ.


Assuntos
Proteína gp120 do Envelope de HIV/fisiologia , HIV-1/imunologia , Macrófagos/imunologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana/fisiologia , Biomarcadores/metabolismo , Diferenciação Celular , Citocinas/biossíntese , Citocinas/imunologia , Regulação da Expressão Gênica , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Proteína gp120 do Envelope de HIV/farmacologia , Infecções por HIV/imunologia , Infecções por HIV/virologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Ativação de Macrófagos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/imunologia , NF-kappa B/genética , NF-kappa B/imunologia , Especificidade de Órgãos , Fenótipo , Cultura Primária de Células , Transdução de Sinais , Produtos do Gene nef do Vírus da Imunodeficiência Humana/farmacologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana/farmacologia
17.
J Cell Physiol ; 227(3): 1090-7, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21567396

RESUMO

The interaction between HIV-1 Nef and the Src kinase Hck in macrophages has been shown to accelerate the progression to AIDS. We previously showed that Nef disturbed the N-glycosylation/trafficking of Fms, a cytokine receptor essential for maintaining macrophages in an anti-inflammatory state, in an Hck-dependent manner. Here, we show the underlying molecular mechanism of this effect. Using various Hck isoforms and their mutants and Golgi-targeting Hck mutants, we confirmed that Hck activation at the Golgi causes the Nef-induced Fms N-glycosylation defect. Importantly, we found that both the co-expression of Nef and Hck and the expression of a Golgi-targeted active Hck mutant caused alterations in the distribution of GM130, a Golgi protein that was shown to be required for efficient protein glycosylation. Moreover, the activation of Hck at the Golgi caused strong serine phosphorylation of the GM130-interacting Golgi structural protein GRASP65, which is known to induce Golgi cisternal unstacking. Using pharmacological inhibitors, we also found that the activation of Hck at the Golgi followed by the activation of the MAP kinase ERK-GRASP65 cascade is involved in the Fms N-glycosylation defect. These results suggest that Nef perturbs the structure and signaling of the Golgi by activating Hck at the Golgi, and thereby, induces the N-glycosylation/trafficking defect of Fms, which is in line with the idea that Src family kinases are crucial Golgi regulators.


Assuntos
Complexo de Golgi/patologia , Complexo de Golgi/virologia , Infecções por HIV/metabolismo , HIV-1/patogenicidade , Proteínas Proto-Oncogênicas c-hck/fisiologia , Transdução de Sinais/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Progressão da Doença , Complexo de Golgi/enzimologia , Células HEK293 , Infecções por HIV/patologia , Infecções por HIV/virologia , Humanos , Transporte Proteico/fisiologia , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo
18.
PLoS One ; 6(11): e27696, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22110726

RESUMO

Nef is a multifunctional HIV-1 protein that accelerates progression to AIDS, and enhances the infectivity of progeny viruses through a mechanism that is not yet understood. Here, we show that the small molecule compound 2c reduces Nef-mediated viral infectivity enhancement. When added to viral producer cells, 2c did not affect the efficiency of viral production itself. However, the infectivity of the viruses produced in the presence of 2c was significantly lower than that of control viruses. Importantly, an inhibitory effect was observed with Nef(+) wild-type viruses, but not with viruses produced in the absence of Nef or in the presence of proline-rich PxxP motif-disrupted Nef, both of which displayed significantly reduced intrinsic infectivity. Meanwhile, the overexpression of the SH3 domain of the tyrosine kinase Hck, which binds to a PxxP motif in Nef, also reduced viral infectivity. Importantly, 2c inhibited Hck SH3-Nef binding, which was more marked when Nef was pre-incubated with 2c prior to its incubation with Hck, indicating that both Hck SH3 and 2c directly bind to Nef and that their binding sites overlap. These results imply that both 2c and the Hck SH3 domain inhibit the interaction of Nef with an unidentified host protein and thereby reduce Nef-mediated infectivity enhancement. The first inhibitory compound 2c is therefore a valuable chemical probe for revealing the underlying molecular mechanism by which Nef enhances the infectivity of HIV-1.


Assuntos
Fármacos Anti-HIV/farmacologia , HIV-1/efeitos dos fármacos , HIV-1/patogenicidade , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Motivos de Aminoácidos , Fármacos Anti-HIV/química , Fármacos Anti-HIV/metabolismo , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , HIV-1/metabolismo , Humanos , Modelos Moleculares , Prolina , Proteínas Proto-Oncogênicas c-hck/química , Proteínas Proto-Oncogênicas c-hck/metabolismo , Produtos do Gene nef do Vírus da Imunodeficiência Humana/química , Domínios de Homologia de src
19.
Exp Parasitol ; 120(4): 306-13, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18786530

RESUMO

Opisthorchis viverrini is the only liver fluke that has been proved to be associated with cholangiocarcinoma (CCA). However, the mechanisms by which O. viverrini participates in the carcinogenesis of CCA are still unclear. To understand the biology and host-parasite interaction related to O. viverrini infection, gene expression profiling of this parasite is required. Here, we constructed the first 5' serial analysis of gene expression (5' SAGE) library of the adult O. viverrini and matched the data with the public data of O. viverrini, Clonorchis sinensis and other related Platyhelminthes and Nematodes. We obtained 12,401 unique tag sequences, of which 6515 (53%) could be matched with the 3419 transcript sequences. The two most abundant tag sequences were vitelline B precursor protein and myoglobin. Often several transcription start sites (TSS) were observed for one transcript. This finding may reflect the dynamic nature of transcriptional initiation events of O. viverrini genes in vivo.


Assuntos
Neoplasias dos Ductos Biliares/parasitologia , Ductos Biliares Intra-Hepáticos , Colangiocarcinoma/parasitologia , Perfilação da Expressão Gênica/métodos , Opistorquíase/parasitologia , Opisthorchis/genética , Animais , Cricetinae , Cyprinidae/parasitologia , Doenças dos Peixes/parasitologia , Interações Hospedeiro-Parasita/genética , Opistorquíase/complicações , Opisthorchis/classificação , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...