Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
1.
PLoS Pathog ; 17(1): e1009183, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33444388

RESUMEN

The antiviral protein kinase R (PKR) is an important host restriction factor, which poxviruses must overcome to productively infect host cells. To inhibit PKR, many poxviruses encode a pseudosubstrate mimic of the alpha subunit of eukaryotic translation initiation factor 2 (eIF2), designated K3 in vaccinia virus. Although the interaction between PKR and eIF2α is highly conserved, some K3 orthologs from host-restricted poxviruses were previously shown to inhibit PKR in a species-specific manner. To better define this host range function, we compared the sensitivity of PKR from 17 mammals to inhibition by K3 orthologs from closely related orthopoxviruses, a genus with a generally broader host range. The K3 orthologs showed species-specific inhibition of PKR and exhibited three distinct inhibition profiles. In some cases, PKR from closely related species showed dramatic differences in their sensitivity to K3 orthologs. Vaccinia virus expressing the camelpox virus K3 ortholog replicated more than three orders of magnitude better in human and sheep cells than a virus expressing vaccinia virus K3, but both viruses replicated comparably well in cow cells. Strikingly, in site-directed mutagenesis experiments between the variola virus and camelpox virus K3 orthologs, we found that different amino acid combinations were necessary to mediate improved or diminished inhibition of PKR derived from different host species. Because there is likely a limited number of possible variations in PKR that affect K3-interactions but still maintain PKR/eIF2α interactions, it is possible that by chance PKR from some potential new hosts may be susceptible to K3-mediated inhibition from a virus it has never previously encountered. We conclude that neither the sensitivity of host proteins to virus inhibition nor the effectiveness of viral immune antagonists can be inferred from their phylogenetic relatedness but must be experimentally determined.


Asunto(s)
Antivirales/antagonistas & inhibidores , Especificidad del Huésped , Orthopoxvirus/clasificación , Orthopoxvirus/fisiología , Infecciones por Poxviridae/virología , Replicación Viral , eIF-2 Quinasa/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Antivirales/metabolismo , Células HeLa , Humanos , Fosforilación , Filogenia , Infecciones por Poxviridae/genética , Infecciones por Poxviridae/metabolismo , Homología de Secuencia , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
2.
J Virol ; 95(6)2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33361422

RESUMEN

DNA damage-inducible transcript 3 (DDIT3) plays important roles in endoplasmic reticulum (ER) stress-induced apoptosis and autophagy, but its role in innate immunity is not clear. Here, we report that DDIT3 inhibits the antiviral immune response during bovine viral diarrhea virus (BVDV) infection by targeting mitochondrial antiviral signaling (MAVS) in Madin-Darby bovine kidney (MDBK) cells and in mice. BVDV infection induced high DDIT3 mRNA and protein expression. DDIT3 overexpression inhibited type I interferon (IFN-I) and IFN-stimulated gene production, thereby promoting BVDV replication, while DDIT3 knockdown promoted the antiviral innate immune response to suppress viral replication. DDIT3 promoted NF-κB-dependent ovarian tumor (OTU) deubiquitinase 1 (OTUD1) expression. Furthermore, OTUD1 induced upregulation of the E3 ubiquitin ligase Smurf1 by deubiquitinating Smurf1, and Smurf1 degraded MAVS in MDBK cells in a ubiquitination-dependent manner, ultimately inhibiting IFN-I production. Moreover, knocking out DDIT3 promoted the antiviral innate immune response to reduce BVDV replication and pathological changes in mice. These findings provide direct insights into the molecular mechanisms by which DDIT3 inhibits IFN-I production by regulating MAVS degradation.IMPORTANCE Extensive studies have demonstrated roles of DDIT3 in apoptosis and autophagy during viral infection. However, the role of DDIT3 in innate immunity remains largely unknown. Here, we show that DDIT3 is positively regulated in bovine viral diarrhea virus (BVDV)-infected Madin-Darby bovine kidney (MDBK) cells and could significantly enhance BVDV replication. Importantly, DDIT3 induced OTU deubiquitinase 1 (OTUD1) expression by activating the NF-κB signaling pathway, thus increasing intracellular Smurf1 protein levels to degrade MAVS and inhibit IFN-I production during BVDV infection. Together, these results indicate that DDIT3 plays critical roles in host innate immunity repression and viral infection facilitation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Virus de la Diarrea Viral Bovina Tipo 1/fisiología , Inmunidad Innata , Factor de Transcripción CHOP/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Replicación Viral , Animales , Antivirales/antagonistas & inhibidores , Antivirales/inmunología , Bovinos , Virus de la Diarrea Viral Bovina Tipo 1/patogenicidad , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Interferón Tipo I/antagonistas & inhibidores , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Ratones , FN-kappa B/metabolismo , Transducción de Señal , Factor de Transcripción CHOP/deficiencia , Factor de Transcripción CHOP/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proteasas Ubiquitina-Específicas/genética , Ubiquitinación
3.
J Virol ; 94(9)2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32102878

RESUMEN

Semen is the primary transmission vehicle for various pathogenic viruses. Initial steps of transmission, including cell attachment and entry, likely occur in the presence of semen. However, the unstable nature of human seminal plasma and its toxic effects on cells in culture limit the ability to study in vitro virus infection and inhibition in this medium. We found that whole semen significantly reduces the potency of antibodies and microbicides that target glycans on the envelope glycoproteins (Envs) of HIV-1. The extraordinarily high concentration of the monosaccharide fructose in semen contributes significantly to the effect by competitively inhibiting the binding of ligands to α1,2-linked mannose residues on Env. Infection and inhibition in whole human seminal plasma are accurately mimicked by a stable synthetic simulant of seminal fluid that we formulated. Our findings indicate that, in addition to the protein content of biological secretions, their small-solute composition impacts the potency of antiviral microbicides and mucosal antibodies.IMPORTANCE Biological secretions allow viruses to spread between individuals. Each type of secretion has a unique composition of proteins, salts, and sugars, which can affect the infectivity potential of the virus and inhibition of this process. Here, we describe HIV-1 infection and inhibition in whole human seminal plasma and a synthetic simulant that we formulated. We discovered that the sugar fructose in semen decreases the activity of a broad and potent class of antiviral agents that target mannose sugars on the envelope protein of HIV-1. This effect of semen fructose likely reduces the efficacy of such inhibitors to prevent the sexual transmission of HIV-1. Our findings suggest that the preclinical evaluation of microbicides and vaccine-elicited antibodies will be improved by their in vitro assessment in synthetic formulations that simulate the effects of semen on HIV-1 infection and inhibition.


Asunto(s)
Fructosa/metabolismo , Fructosa/farmacología , Semen/metabolismo , Adulto , Antiinfecciosos/farmacología , Antivirales/antagonistas & inhibidores , Antivirales/farmacología , Línea Celular Tumoral , Productos del Gen env/metabolismo , Genes env/genética , Células HEK293 , Infecciones por VIH/virología , VIH-1/inmunología , Humanos , Masculino , Manosa/metabolismo , Polisacáridos/inmunología , Polisacáridos/metabolismo , Semen/virología , Productos del Gen env del Virus de la Inmunodeficiencia Humana/metabolismo
4.
J Virol ; 94(23)2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-32938761

RESUMEN

SARS-CoV-2, a novel coronavirus (CoV) that causes COVID-19, has recently emerged causing an ongoing outbreak of viral pneumonia around the world. While distinct from SARS-CoV, both group 2B CoVs share similar genome organization, origins to bat CoVs, and an arsenal of immune antagonists. In this report, we evaluate type I interferon (IFN-I) sensitivity of SARS-CoV-2 relative to the original SARS-CoV. Our results indicate that while SARS-CoV-2 maintains similar viral replication to SARS-CoV, the novel CoV is much more sensitive to IFN-I. In Vero E6 and in Calu3 cells, SARS-CoV-2 is substantially attenuated in the context of IFN-I pretreatment, whereas SARS-CoV is not. In line with these findings, SARS-CoV-2 fails to counteract phosphorylation of STAT1 and expression of ISG proteins, while SARS-CoV is able to suppress both. Comparing SARS-CoV-2 and influenza A virus in human airway epithelial cultures, we observe the absence of IFN-I stimulation by SARS-CoV-2 alone but detect the failure to counteract STAT1 phosphorylation upon IFN-I pretreatment, resulting in near ablation of SARS-CoV-2 infection. Next, we evaluated IFN-I treatment postinfection and found that SARS-CoV-2 was sensitive even after establishing infection. Finally, we examined homology between SARS-CoV and SARS-CoV-2 in viral proteins shown to be interferon antagonists. The absence of an equivalent open reading frame 3b (ORF3b) and genetic differences versus ORF6 suggest that the two key IFN-I antagonists may not maintain equivalent function in SARS-CoV-2. Together, the results identify key differences in susceptibility to IFN-I responses between SARS-CoV and SARS-CoV-2 that may help inform disease progression, treatment options, and animal model development.IMPORTANCE With the ongoing outbreak of COVID-19, differences between SARS-CoV-2 and the original SARS-CoV could be leveraged to inform disease progression and eventual treatment options. In addition, these findings could have key implications for animal model development as well as further research into how SARS-CoV-2 modulates the type I IFN response early during infection.


Asunto(s)
Antivirales/farmacología , Betacoronavirus/efectos de los fármacos , Interferón Tipo I/farmacología , Interferón-alfa/farmacología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Animales , Antivirales/antagonistas & inhibidores , Antivirales/metabolismo , Betacoronavirus/inmunología , Betacoronavirus/fisiología , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Humanos , Interferón Tipo I/antagonistas & inhibidores , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Interferón-alfa/antagonistas & inhibidores , Interferón-alfa/inmunología , Interferón-alfa/metabolismo , Fosforilación , Proteínas Recombinantes/farmacología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , SARS-CoV-2 , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Células Vero , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral/efectos de los fármacos
5.
Int J Mol Sci ; 22(7)2021 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-33806254

RESUMEN

The global COVID-19 pandemic caused by SARS-CoV-2 has resulted in over 2.2 million deaths. Disease outcomes range from asymptomatic to severe with, so far, minimal genotypic change to the virus so understanding the host response is paramount. Transcriptomics has become incredibly important in understanding host-pathogen interactions; however, post-transcriptional regulation plays an important role in infection and immunity through translation and mRNA stability, allowing tight control over potent host responses by both the host and the invading virus. Here, we apply ribosome profiling to assess post-transcriptional regulation of host genes during SARS-CoV-2 infection of a human lung epithelial cell line (Calu-3). We have identified numerous transcription factors (JUN, ZBTB20, ATF3, HIVEP2 and EGR1) as well as select antiviral cytokine genes, namely IFNB1, IFNL1,2 and 3, IL-6 and CCL5, that are restricted at the post-transcriptional level by SARS-CoV-2 infection and discuss the impact this would have on the host response to infection. This early phase restriction of antiviral transcripts in the lungs may allow high viral load and consequent immune dysregulation typically seen in SARS-CoV-2 infection.


Asunto(s)
Citocinas/genética , Procesamiento Postranscripcional del ARN , Ribosomas/metabolismo , Ribosomas/virología , SARS-CoV-2/inmunología , Factores de Transcripción/genética , Animales , Antivirales/antagonistas & inhibidores , Línea Celular Tumoral , Chlorocebus aethiops , Biología Computacional , Citocinas/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/virología , Perfilación de la Expresión Génica , Interacciones Microbiota-Huesped , Humanos , Inmunidad Innata/genética , Pulmón/inmunología , Pulmón/virología , ARN Mensajero/metabolismo , RNA-Seq , Ribosomas/genética , SARS-CoV-2/metabolismo , Factores de Transcripción/metabolismo , Transcriptoma , Células Vero
6.
J Med Virol ; 91(5): 803-812, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30570770

RESUMEN

Interleukin 6 (IL-6) is a pleiotropic cytokine with pivotal functions in the regulation of the biological responses of several target cells, including hepatocytes. Previous studies have shown that serum IL-6 levels are increased in hepatitis B patients. However, the role of IL-6 in modulating the anti-hepatitis B virus (HBV) activity of interferon-α (IFN-α) remains unclear. In this study, we found that both HBV and viral proteins could induce the expression of IL-6 in hepatocytes (LO2 and HepG2). Exogenous IL-6 had no effect on HBV replication, whereas knockdown of IL-6 expression by RNAi inhibited that. Interestingly, IFN-α markedly induced IL-6 expression in hepatocytes, especially in HBV replicating hepatocytes. In turn, IL-6 impaired the anti-HBV efficiency of IFN-α by decreases the expression of IFN-α downstream effectors by upregulation of suppressor of cytokine signaling-3 (SOCS3). Furthermore, we demonstrated that downregulation of SOCS3 improved IFN antiviral activity to some extent in HBV replicating hepatocytes. These data provided new insights for a better understanding of the mechanism of IFN-α resistance and may represent a novel therapeutic strategy to efficiently target HBV infection.


Asunto(s)
Antivirales/antagonistas & inhibidores , Virus de la Hepatitis B/inmunología , Interacciones Huésped-Patógeno , Evasión Inmune , Interferón-alfa/antagonistas & inhibidores , Interleucina-6/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Línea Celular , Hepatocitos/virología , Humanos
7.
J Dairy Sci ; 102(11): 9559-9569, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31495632

RESUMEN

Antiviral neuraminidase inhibitors, such as oseltamivir, zanamivir, and peramivir, are widely used for treatment of influenza virus infection. We reported previously that oseltamivir inhibits the viral growth cycle, ameliorates symptoms, and reduces viral antigen quantities. Suppressed viral antigen production, however, induces a reduction of acquired antiviral humoral immunity, and increases the incidence of re-infection rate in the following year. To achieve effective treatment of influenza virus infection, it is necessary to overcome these adverse effects of antiviral neuraminidase inhibitors. Feeding of yogurt fermented with Lactobacillus delbrueckii ssp. bulgaricus (L. bulgaricus) OLL1073R-1 is reported to have immune-stimulatory effects on influenza virus infection in mice and humans. In the present study, we assessed the effect of feeding L. bulgaricus OLL1073R-1 yogurt cultures (YC) on local and systemic humoral immune responses, which were suppressed by oseltamivir treatment, in mice infected with influenza A virus. Yogurt culture (1.14 × 108 cfu/0.4 mL per mouse per day) or sterile water (vehicle) was administered by intragastric gavage for 35 d. At d 22, influenza A virus/Puerto Rico/8/34 (H1N1) (PR8; 0.5 pfu/15 µL per mouse) was instilled intranasally, followed immediately by oral administration of oseltamivir (50 µg/100 µL per mouse, twice daily) or 5% methylcellulose (100 µL/mouse) as a vehicle for 13 d. Titers of anti-PR8-specific IgG and IgA in serum and mucosal secretory IgA (S-IgA) and IgG in bronchoalveolar lavage fluid (BALF) were analyzed by ELISA at 14 d after infection. Oseltamivir significantly suppressed the induction of anti-PR8-specific IgG and IgA in serum and S-IgA and IgG in BALF after infection. Feeding YC mildly but significantly stimulated production of PR8-specific IgA in serum, S-IgA in BALF, and IgG in serum without changing the IgG2a:IgG1 ratio. We analyzed the neutralizing activities against PR8 in serum and BALF and found that oseltamivir also reduced protective immunity, and YC feeding abrogated this effect. The immune-stimulatory tendency of YC on anti-PR8-specific IgA and IgG titers in serum and BALF was also detected in mice re-infected with PR8, but the effect was insignificant, unlike the effect of YC in the initial infection.


Asunto(s)
Antivirales/uso terapéutico , Inmunidad Humoral/efectos de los fármacos , Lactobacillus delbrueckii , Neuraminidasa/antagonistas & inhibidores , Infecciones por Orthomyxoviridae/inmunología , Oseltamivir/uso terapéutico , Probióticos/uso terapéutico , Proteínas Virales/antagonistas & inhibidores , Alimentación Animal , Animales , Antivirales/efectos adversos , Antivirales/antagonistas & inhibidores , Inhibidores Enzimáticos/uso terapéutico , Femenino , Humanos , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Lactobacillus delbrueckii/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/dietoterapia , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Oseltamivir/efectos adversos , Oseltamivir/antagonistas & inhibidores , Yogur
8.
Microbiol Immunol ; 62(5): 341-347, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29603339

RESUMEN

Treatment of human embryonic lung fibroblast (HEL) cells with tricin (4', 5, 7-trihydroxy-3', 5'-dimethoxyflavone) following infection with human cytomegalovirus (HCMV) reportedly significantly suppresses HCMV replication. In the present work, the mechanisms for the anti-HCMV effects of tricin in HEL cells were examined. It was found that exposure of HEL cells to tricin inhibited HCMV replication, with concomitant decreases in amounts of transcripts of the CC chemokine RANTES (CCL5)-encoding gene and in expression of the CCL5 protein. It was also found that transcripts of HCMV immediate early 1 (IE1), and HCMV UL54 (encoding DNA polymerase) and replication of HCMV was significantly lower in CCL5 gene-knockdown cells. These results suggest that the anti-HCMV activity of tricin differs from that of ganciclovir and that CCL5 is one of the chemokines involved in HCMV replication. In addition, it is possible that chemokine CCL5 is one of the targets of tricin.


Asunto(s)
Antivirales/antagonistas & inhibidores , Quimiocina CCL5/genética , Infecciones por Citomegalovirus/virología , Citomegalovirus/crecimiento & desarrollo , Flavonoides/antagonistas & inhibidores , Expresión Génica/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Línea Celular , Citomegalovirus/efectos de los fármacos , Citomegalovirus/genética , Replicación del ADN/efectos de los fármacos , ADN Polimerasa Dirigida por ADN , Fibroblastos/efectos de los fármacos , Ganciclovir/antagonistas & inhibidores , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , Proteínas Inmediatas-Precoces , ARN Interferente Pequeño , Transfección , Proteínas Virales/genética
9.
J Virol ; 90(18): 8281-92, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27384656

RESUMEN

UNLABELLED: Porcine epidemic diarrhea virus (PEDV) is a worldwide-distributed alphacoronavirus, but the pathogenesis of PEDV infection is not fully characterized. During virus infection, type I interferon (IFN) is a key mediator of innate antiviral responses. Most coronaviruses develop some strategy for at least partially circumventing the IFN response by limiting the production of IFN and by delaying the activation of the IFN response. However, the molecular mechanisms by which PEDV antagonizes the antiviral effects of interferon have not been fully characterized. Especially, how PEDV impacts IFN signaling components has yet to be elucidated. In this study, we observed that PEDV was relatively resistant to treatment with type I IFN. Western blot analysis showed that STAT1 expression was markedly reduced in PEDV-infected cells and that this reduction was not due to inhibition of STAT1 transcription. STAT1 downregulation was blocked by a proteasome inhibitor but not by an autophagy inhibitor, strongly implicating the ubiquitin-proteasome targeting degradation system. Since PEDV infection-induced STAT1 degradation was evident in cells pretreated with the general tyrosine kinase inhibitor, we conclude that STAT1 degradation is independent of the IFN signaling pathway. Furthermore, we report that PEDV-induced STAT1 degradation inhibits IFN-α signal transduction pathways. Pharmacological inhibition of STAT1 degradation rescued the ability of the host to suppress virus replication. Collectively, these data show that PEDV is capable of subverting the type I interferon response by inducing STAT1 degradation. IMPORTANCE: In this study, we show that PEDV is resistant to the antiviral effect of IFN. The molecular mechanism is the degradation of STAT1 by PEDV infection in a proteasome-dependent manner. This PEDV infection-induced STAT1 degradation contributes to PEDV replication. Our findings reveal a new mechanism evolved by PEDV to circumvent the host antiviral response.


Asunto(s)
Antivirales/antagonistas & inhibidores , Interacciones Huésped-Patógeno , Evasión Inmune , Interferón-alfa/antagonistas & inhibidores , Virus de la Diarrea Epidémica Porcina/patogenicidad , Factor de Transcripción STAT1/antagonistas & inhibidores , Animales , Western Blotting , Línea Celular , Chlorocebus aethiops , Infecciones por Coronaviridae , Regulación hacia Abajo , Proteolisis , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Porcinos
10.
Virol J ; 14(1): 215, 2017 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-29110727

RESUMEN

BACKGROUND: Interferon (IFN) inhibits viruses by inducing several hundred cellular genes, aptly named 'interferon (IFN)-stimulated genes' (ISGs). The only two RNA viruses of the Pneumovirus genus of the Paramyxoviridae family, namely Respiratory Syncytial Virus (RSV) and Pneumonia Virus of Mice (PVM), each encode two nonstructural (NS) proteins that share no sequence similarity but yet suppress IFN. Since suppression of IFN underlies the ability of these viruses to replicate in the host cells, the mechanism of such suppression has become an important area of research. This Short Report is an important extension of our previous efforts in defining this mechanism. RESULTS: We show that, like their PVM counterparts, the RSV NS proteins also target multiple members of the ISG family. While significantly extending the substrate repertoire of the RSV NS proteins, these results, unexpectedly, also reveal that the target preferences of the NS proteins of the two viruses are entirely different. This is surprising since the two Pneumoviruses are phylogenetically close with similar genome organization and gene function, and the NS proteins of both also serve as suppressors of host IFN response. CONCLUSION: The finding that the NS proteins of the two highly similar viruses suppress entirely different members of the ISG family raises intriguing questions of pneumoviral NS evolution and mechanism of action.


Asunto(s)
Virus de la Neumonía Murina/fisiología , Virus Sincitial Respiratorio Humano/fisiología , Proteínas no Estructurales Virales/metabolismo , Animales , Antivirales/antagonistas & inhibidores , Variación Genética , Células HEK293 , Interacciones Huésped-Patógeno/genética , Humanos , Interferones/antagonistas & inhibidores , Ratones , Especificidad por Sustrato , Proteínas no Estructurales Virales/genética
11.
Microbiol Immunol ; 61(6): 232-238, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28463406

RESUMEN

JC polyomavirus (JCPyV) is the causative agent of the demyelinating disease of the central nervous system known as progressive multifocal leukoencephalopathy (PML), which occurs in immunocompromised patients. Moreover, patients treated with natalizumab for multiple sclerosis or Crohn disease can develop PML, which is then termed natalizumab-related PML. Because few drugs are currently available for treating PML, many antiviral agents are being investigated. It has been demonstrated that the topoisomerase I inhibitors topotecan and ß-lapachone have inhibitory effects on JCPyV replication in IMR-32 cells. However, both of these drugs have marginal inhibitory effects on virus propagation in JC1 cells according to RT-PCR analysis. In the present study, the inhibitory effect of another topoisomerase I inhibitor, 7-ethy-10-[4-(1-piperidino)-1-piperidino] carbonyloxy camptothecin (CPT11), was assessed by investigating viral replication, propagation, and viral protein 1 (VP1) production in cultured cells. JCPyV replication was assayed using real-time PCR combined with Dpn I treatment in IMR-32 cells transfected with JCPyV DNA. It was found that JCPyV replicates less in IMR-32 cells treated with CPT11 than in untreated cells. Moreover, CPT11 treatment of JCI cells persistently infected with JCPyV led to a dose-dependent reduction in JCPyV DNA and VP1 production. Additionally, the inhibitory effect of CPT11 was found to be stronger than those of topotecan and ß-lapachone. These findings suggest that CPT11 may be a potential anti-JCPyV agent that could be used to treat PML.


Asunto(s)
Antivirales/antagonistas & inhibidores , Camptotecina/antagonistas & inhibidores , Virus JC/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Camptotecina/administración & dosificación , Camptotecina/toxicidad , Línea Celular/efectos de los fármacos , Línea Celular/virología , Proliferación Celular/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , ADN Viral/genética , Relación Dosis-Respuesta a Droga , Humanos , Concentración 50 Inhibidora , Virus JC/genética , Leucoencefalopatía Multifocal Progresiva/tratamiento farmacológico , Naftoquinonas/antagonistas & inhibidores , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Inhibidores de Topoisomerasa I/farmacología , Topotecan/antagonistas & inhibidores , Proteínas Virales/efectos de los fármacos
12.
Cell Microbiol ; 15(10): 1688-706, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23522008

RESUMEN

Flavivirus capsid (C) protein is a key structural component of virus particles. The non-structural role of C protein in the pathogenesis of arthropod-borne flaviviruses is not clearly deciphered. This study showed that West Nile virus (WNV) and dengue virus (DENV) utilized C protein to reduce human Sec3p (hSec3p) levels at post-transcriptional level through activation of chymotrypsin-like proteolytic function of 20S proteasome. Mutagenesis studies confirmed amino acids 14, 109-114 of WNV C protein and 13, 102-107 of DENV C protein played an important role in activating the proteolytic function of 20S proteasome. Amino acid residues at 14 (WNV) and 13 (DENV) of C protein were important for C protein-hSec3p binding and physical interaction between C protein and hSec3p was essential to execute hSec3p degradation. Degradation motif required to degrade hSec3p resided between amino acid residues 109-114 of WNV C protein and 102-107 of DENV C protein. Proteasomes, hSec3p binding motif and degradation motif on C protein must be intact for efficient flavivirus production. Clinical isolates of DENV showed more pronounced effect in manipulating the proteasomes and reducing hSec3p levels. This study portrayed the non-structural function of C protein that helped the flavivirus to nullify the antiviral activity of hSec3p by accelerating its degradation and facilitating efficient binding of elongation factor 1α with flaviviral RNA genome.


Asunto(s)
Antivirales/antagonistas & inhibidores , Proteínas de la Cápside/metabolismo , Virus del Dengue/fisiología , Interacciones Huésped-Patógeno , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas de Transporte Vesicular/antagonistas & inhibidores , Virus del Nilo Occidental/fisiología , Proteínas de la Cápside/genética , Línea Celular , Análisis Mutacional de ADN , Humanos , Modelos Biológicos , Unión Proteica , Mapeo de Interacción de Proteínas , Proteolisis
13.
Br J Nutr ; 111(9): 1586-93, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24495389

RESUMEN

Our previous studies have shown that vitamin A (VA) status is associated with antiviral immunity and pathogenic conditions in enterovirus 71 (EV71)-infected children. In the present study, we established an in vitro model to investigate the effects and potential mechanism of the antiviral activity of VA. Human monocytic U937 cells were cultured in vitro and infected with EV71. All-trans-retinoic acid (ATRA), the active metabolite of VA, and Ro 41-5253, a retinoic acid receptor-α (RAR-α) antagonist, were used as the experimental treatment agents. The percentage of EV71-infected cells and apoptosis induced by EV71 were determined using flow cytometry. The level of interferon-α (IFN-α) in the supernatants of the cultures was detected using ELISA. The expression of retinoid-induced gene I (RIG-I) and its downstream genes was examined with real-time quantitative PCR. The results indicated that ATRA reduced the percentage of EV71-infected cells and protected cells against EV71-induced apoptosis. Correspondingly, ATRA increased the production of IFN-α one of the most important antiviral cytokines, at both mRNA and protein levels in EV71-infected cells. In addition, the expression of RIG-I mRNA and its downstream genes was up-regulated by ATRA in EV71-infected cells. Ro 41-5253 abrogated the inhibitory effects of ATRA on EV71. The present findings suggest that ATRA is an interferon-inducing agent with antiviral activity against EV71 in vitro and that its actions are mediated at least in part by RAR-α activity and the RIG-I signalling pathway.


Asunto(s)
Antivirales/farmacología , Enterovirus Humano A/efectos de los fármacos , Monocitos/efectos de los fármacos , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal/efectos de los fármacos , Tretinoina/farmacología , Antivirales/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Benzoatos/farmacología , Línea Celular , Cromanos/farmacología , Enterovirus Humano A/crecimiento & desarrollo , Enterovirus Humano A/inmunología , Enterovirus Humano A/metabolismo , Infecciones por Enterovirus/tratamiento farmacológico , Infecciones por Enterovirus/inmunología , Infecciones por Enterovirus/metabolismo , Infecciones por Enterovirus/microbiología , Antagonistas de Hormonas/farmacología , Humanos , Inmunidad Innata/efectos de los fármacos , Interferón-alfa/genética , Interferón-alfa/metabolismo , Viabilidad Microbiana/efectos de los fármacos , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/virología , ARN Mensajero/metabolismo , Receptor de Interferón alfa y beta/biosíntesis , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Receptores de Ácido Retinoico/antagonistas & inhibidores , Receptores de Ácido Retinoico/biosíntesis , Receptores de Ácido Retinoico/química , Receptores de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico , Tretinoina/antagonistas & inhibidores , Regulación hacia Arriba/efectos de los fármacos , Proteínas Virales/metabolismo
14.
J Immunol ; 188(5): 2111-7, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22271650

RESUMEN

The manipulation of signals downstream of the TCR can have profound consequences for T cell development, function, and homeostasis. Diacylglycerol (DAG) produced after TCR stimulation functions as a secondary messenger and mediates the signaling to Ras-MEK-Erk and NF-κB pathways in T cells. DAG kinases (DGKs) convert DAG into phosphatidic acid, resulting in termination of DAG signaling. In this study, we demonstrate that DAG metabolism by DGKs can serve a crucial function in viral clearance upon lymphocytic choriomeningitis virus infection. Ag-specific CD8(+) T cells from DGKα(-/-) and DGKζ(-/-) mice show enhanced expansion and increased cytokine production after lymphocytic choriomeningitis virus infection, yet DGK-deficient memory CD8(+) T cells exhibit impaired expansion after rechallenge. Thus, DGK activity plays opposing roles in the expansion of CD8(+) T cells during the primary and memory phases of the immune response, whereas consistently inhibiting antiviral cytokine production.


Asunto(s)
Linfocitos T CD8-positivos/enzimología , Linfocitos T CD8-positivos/inmunología , Diacilglicerol Quinasa/fisiología , Memoria Inmunológica , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/patología , Animales , Antivirales/antagonistas & inhibidores , Antivirales/metabolismo , Linfocitos T CD8-positivos/virología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Línea Celular , Células Clonales , Cricetinae , Citocinas/antagonistas & inhibidores , Citocinas/biosíntesis , Diacilglicerol Quinasa/deficiencia , Diacilglicerol Quinasa/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Epítopos de Linfocito T/inmunología , Memoria Inmunológica/genética , Coriomeningitis Linfocítica/genética , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Noqueados , Ratones Transgénicos
15.
J Biol Regul Homeost Agents ; 27(4): 1001-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24382181

RESUMEN

Type III interferons (IFN-lambda) are the most recently discovered members of IFN family. Synergism between different IFN types is well established, but for type I and type III IFNs no conclusive evidence has been reported so far. Possible synergism/antagonism between IFN-alpha and IFN-lambda in the inhibition of virus replication (EMCV, WNV lineage 1 and 2, CHIKV and HSV-1), and in the activation of intracellular pathways of IFN response (MxA and 2'-5' OAS) was evaluated in different cell lines (Vero E6, A549 and Wish cells). The antiviral potency of IFN-lambda1 and -l2 was lower than that of IFN-alpha. When IFN-alpha and -lambda were used together, the Combination Index (CI) for virus inhibition was greater than 1 virtually for all virus/host cell systems, indicating antagonistic effect. Antagonism between IFN-alpha and -l was also observed for the induction of mRNA for both MxA and 2'-5'OAS. Elucidating the interplay between IFN-alpha and -lambda may help to better understand innate defence mechanisms against viral infections, including the molecular mechanisms underlying the influence of IL-28B polymorphisms in the response to HCV and other viral infections.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/genética , Antivirales/antagonistas & inhibidores , Interferón-alfa/antagonistas & inhibidores , Interferones/farmacología , Interleucinas/farmacología , Animales , Chlorocebus aethiops , Virus de la Encefalomiocarditis/efectos de los fármacos , Humanos , Proteínas de Resistencia a Mixovirus/genética , Células Vero , Replicación Viral/efectos de los fármacos
16.
Mutat Res ; 752(1-2): 14-20, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23402882

RESUMEN

Ribavirin (1-ß-d-ribofuranosyl-1,2,4-triazole-3-carboxamide) is a widely used broad-spectrum antiviral drug. Recently, several reports revealed genotoxic effects of ribavirin in vivo and in vitro, which were correlated with the production of reactive oxygen species (ROS). This study aimed to evaluate the genotoxicity of ribavirin and to investigate the role of the natural antioxidant silymarin to modulate this genotoxicity. Male albino mice (age, 8-10 weeks) were injected intraperitoneally (i.p.) with ribavirin at three dose levels (20, 75 and 130 mg/kg bw) either in a single injection (acute treatment) or in multiple injections on five consecutive days (sub-acute treatment). Other comparable groups were treated with silymarin (70 mg/kg bw) 1h before the injection with ribavirin. Mice were sacrificed at different sampling times (24, 48 and 72 h) after the last ribavirin treatment. Micronucleus (MN) and single-strand conformation polymorphism (SSCP) assays were used to assess genotoxic and cytotoxic effects of ribavirin and to evaluate the protective effect of the pre-treatment with silymarin. Our results reveal genotoxic and cytotoxic effects of ribavirin in the MN assay. Pre-treatment with silymarin reduced the toxicity of ribavirin. In the SSCP assay, ribavirin treatment did not induce any mutations in the two selected sites in the D-loop of mitochondrial DNA (mtDNA).


Asunto(s)
Antioxidantes/farmacología , Antivirales/toxicidad , Ribavirina/toxicidad , Silimarina/farmacología , Animales , Antivirales/antagonistas & inhibidores , Masculino , Ratones , Pruebas de Micronúcleos , Pruebas de Mutagenicidad , Mutación/efectos de los fármacos , Polimorfismo Conformacional Retorcido-Simple , Ribavirina/antagonistas & inhibidores
17.
J Immunol ; 184(9): 5179-85, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20308629

RESUMEN

Viperin is an antiviral protein whose expression is highly upregulated during viral infections via IFN-dependent and/or IFN-independent pathways. We examined the molecular alterations induced by the transcriptional activator IFN regulatory factor (IRF)-1 and found viperin to be among the group of IRF-1 regulated genes. From these data, it was not possible to distinguish genes that are primary targets of IRF-1 and those that are targets of IRF-1-induced proteins, like IFN-beta. In this study, we show that IRF-1 directly binds to the murine viperin promoter to the two proximal IRF elements and thereby induces viperin expression. Infection studies with embryonal fibroblasts from different gene knock-out mice demonstrate that IRF-1 is essential, whereas the type I IFN system is dispensable for vesicular stomatitis virus induced viperin gene transcription. Further, IRF-1, but not IFN type I, mediates the induction of viperin transcription after IFN-gamma treatment. In contrast, IRF-1 is not required for IFN-independent viperin induction by Newcastle disease virus infection and by infection with a vesicular stomatitis virus mutant that is unable to block IFN expression and secretion. We conclude that the IRF-1 mediated type I IFN independent mechanism of enhanced viperin expression provides a redundant mechanism to protect cells from viral infections. This mechanism becomes important when viruses evade innate immunity by antagonizing the induction and function of the IFN system.


Asunto(s)
Antivirales/farmacología , Factor 1 Regulador del Interferón/fisiología , Interferón Tipo I/fisiología , Proteínas/genética , Activación Transcripcional/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Animales , Antivirales/antagonistas & inhibidores , Células Cultivadas , Factor 1 Regulador del Interferón/deficiencia , Factor 1 Regulador del Interferón/genética , Interferón Tipo I/antagonistas & inhibidores , Ratones , Ratones Noqueados , Células 3T3 NIH , Regiones Promotoras Genéticas/inmunología , Proteínas/metabolismo , Transducción de Señal/inmunología , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología , Estomatitis Vesicular/inmunología , Estomatitis Vesicular/prevención & control , Replicación Viral/inmunología
18.
Nat Med ; 4(12): 1397-400, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9846577

RESUMEN

Animal cells have developed many ways to suppress viral replication, and viruses have evolved diverse strategies to resist these. Here we provide evidence that the virion infectivity factor protein of human immunodeficiency virus type 1 (HIV-1) functions to counteract a newly discovered activity in human cells that otherwise inhibits virus replication. This anti-viral phenotype is shown by human T cells, the principal in vivo targets for HIV-1, and, based on our present understanding of virion infectivity factor action, is presumed to act by interfering with a late step(s) in the virus life cycle. These observations indicate that the inhibition of virion infectivity factor function in vivo may prevent HIV-1 replication by 'unmasking' an innate anti-viral phenotype.


Asunto(s)
Antivirales/antagonistas & inhibidores , VIH-1/patogenicidad , Proteínas Reguladoras y Accesorias Virales/fisiología , Animales , Células Cultivadas , Productos del Gen vif/fisiología , VIH-1/fisiología , Humanos , Inmunidad Innata , Fenotipo , Linfocitos T/fisiología , Linfocitos T/virología , Replicación Viral , Productos del Gen vif del Virus de la Inmunodeficiencia Humana
19.
Antiviral Res ; 187: 105018, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33476709

RESUMEN

MBX-2168 has a mechanism of action similar to that of acyclovir (ACV) and ganciclovir (GCV), but two unique steps differentiate this drug from ACV/GCV. First, MBX-2168 is, at least partially, phosphorylated by the endogenous cellular kinase TAOK3 to a monophosphate. The second involves the removal of a moiety at the 6-position of MBX-2168-MP by adenosine deaminase like protein-1 (ADAL-1). It has been previously demonstrated that co-incubation with pentostatin (dCF), an ADAL-1 inhibitor, antagonizes the anti-viral activity of MBX-2168. We therefore hypothesize that inhibiting ADAL-1 results in a reduction of active compound produced in virus-infected cells. To test this, we examined the effect dCF has on the conversion of MBX-2168 to a triphosphate in HSV-1 and HCMV-infected cells. Our results demonstrate incubation of MBX-2168 alone or with dCF in HCMV-infected cells resulted in 53.1 ± 0.7 and 39.4 ± 1.5 pmol triphosphate/106 cells at 120 h, respectively. Incubation of MBX-2168 alone or with dCF in Vero cells resulted in 12.8 ± 0.1 and 6.7 ± 0.7 pmol triphosphate/106 cells at 24 h, respectively. HSV-1-infected Vero cells demonstrated no statistical difference in triphosphate accumulation at 24 h (13.1 ± 0.3 pmol triphosphate/106 cells). As expected, incubation with dCF resulted in the accumulation of MBX-2168-MP in both HFF (9.8 ± 0.9 pmol MBX-2168-MP/106 cells at 120 h) and Vero cells (4.7 ± 0.3 pmol MBX-2168-MP/106 cells at 24 h) while no detectable levels of monophosphate were observed in cultures not incubated with dCF. We conclude that dCF antagonizes the anti-viral effect of MBX-2168 by inhibiting the production of triphosphate, the active compound.


Asunto(s)
Antivirales/antagonistas & inhibidores , Antivirales/farmacología , Ciclopropanos/antagonistas & inhibidores , Citomegalovirus/efectos de los fármacos , Guanina/análogos & derivados , Herpesvirus Humano 1/efectos de los fármacos , Pentostatina/farmacología , Polifosfatos/metabolismo , Aciclovir/farmacología , Animales , Línea Celular , Chlorocebus aethiops , Ciclopropanos/farmacología , Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/virología , Fibroblastos/virología , Prepucio/citología , Ganciclovir/farmacología , Guanina/antagonistas & inhibidores , Guanina/farmacología , Herpes Simple/tratamiento farmacológico , Herpes Simple/virología , Interacciones Microbiota-Huesped , Humanos , Mutación con Pérdida de Función , Masculino , Fosforilación , Células Vero , Replicación Viral/efectos de los fármacos
20.
Biochem Pharmacol ; 186: 114436, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33539815

RESUMEN

Thrombocytopenia is common among patients with viral hepatitis, limiting the use of antiviral therapy. Eltrombopag (EP) is a thrombopoietin receptor (TPO-R) agonist that has been approved for treatment of immune thrombocytopenia patients with hepatitis virus infection. Interferon-α (IFN-α) plays a crucial role in the antiviral response, and is recommended as the first-line agent for chronic hepatitis B patients. Here, we investigated whether EP inhibits the production of IFN-stimulated genes (ISGs) induced by IFN-α through the TPO-R-independent pathway by mediating reactive oxygen species production by iron chelation. Our results assessed the inhibitory effect of EP on IFN-α signaling, which contributes to the downregulation of ISGs produced by monocytes and sheds light on the underlying mechanisms using iron chelation to treat patients with hepatitis-related immunological thrombocytopenia.


Asunto(s)
Antivirales/metabolismo , Benzoatos/farmacología , Hidrazinas/farmacología , Interferón-alfa/metabolismo , Hierro/metabolismo , Leucocitos Mononucleares/metabolismo , Pirazoles/farmacología , Adulto , Animales , Antivirales/antagonistas & inhibidores , Benzoatos/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Femenino , Hepatitis B Crónica/tratamiento farmacológico , Hepatitis B Crónica/metabolismo , Humanos , Hidrazinas/uso terapéutico , Interferón-alfa/antagonistas & inhibidores , Leucocitos Mononucleares/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Pirazoles/uso terapéutico , Receptores de Trombopoyetina/agonistas , Receptores de Trombopoyetina/metabolismo , Células THP-1/efectos de los fármacos , Células THP-1/metabolismo , Trombocitopenia/tratamiento farmacológico , Trombocitopenia/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda