Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Molecules ; 26(23)2021 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-34885710

RESUMEN

Structural and biochemical studies elucidate that PAN may contribute to the host protein shutdown observed during influenza A infection. Thus, inhibition of the endonuclease activity of viral RdRP is an attractive approach for novel antiviral therapy. In order to envisage structurally diverse novel compounds with better efficacy as PAN endonuclease inhibitors, a ligand-based-pharmacophore model was developed using 3D-QSAR pharmacophore generation (HypoGen algorithm) methodology in Discovery Studio. As the training set, 25 compounds were taken to generate a significant pharmacophore model. The selected pharmacophore Hypo1 was further validated by 12 compounds in the test set and was used as a query model for further screening of 1916 compounds containing 71 HIV-1 integrase inhibitors, 37 antibacterial inhibitors, 131 antiviral inhibitors and other 1677 approved drugs by the FDA. Then, six compounds (Hit01-Hit06) with estimated activity values less than 10 µM were subjected to ADMET study and toxicity assessment. Only one potential inhibitory 'hit' molecule (Hit01, raltegravir's derivative) was further scrutinized by molecular docking analysis on the active site of PAN endonuclease (PDB ID: 6E6W). Hit01 was utilized for designing novel potential PAN endonuclease inhibitors through lead optimization, and then compounds were screened by pharmacophore Hypo1 and docking studies. Six raltegravir's derivatives with significant estimated activity values and docking scores were obtained. Further, these results certainly do not confirm or indicate the seven compounds (Hit01, Hit07, Hit08, Hit09, Hit10, Hit11 and Hit12) have antiviral activity, and extensive wet-laboratory experimentation is needed to transmute these compounds into clinical drugs.


Asunto(s)
Adenosina Trifosfatasas/química , Endonucleasas/química , Inhibidores Enzimáticos/química , Gripe Humana/enzimología , Adenosina Trifosfatasas/antagonistas & inhibidores , Adenosina Trifosfatasas/ultraestructura , Dominio Catalítico/efectos de los fármacos , Diseño de Fármacos/tendencias , Endonucleasas/antagonistas & inhibidores , Endonucleasas/ultraestructura , Humanos , Gripe Humana/tratamiento farmacológico , Gripe Humana/virología , Ligandos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad Cuantitativa
2.
Curr Top Med Chem ; 20(23): 2046-2055, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31738149

RESUMEN

BACKGROUND: Neuraminidase (NA), a major glycoprotein found on the surface of the influenza virus, is an important target for the prophylaxis and treatment of influenza virus infections. Recently, several plant-derived polyphenols, especially caffeic acid analogs, have been reported to exert the inhibitory activity against NA. OBJECTIVE: Herein, we aimed to investigate the anti-influenza NA activity of caffeic acid and its hydroxycinnamate analogues, rosmarinic acid and salvianolic acid A, in comparison to a known NA inhibitor, oseltamivir. METHODS: In vitro MUNANA-based NA inhibitory assay was used to evaluate the inhibitory activity of the three interested hydroxycinnamic compounds towards the influenza NA enzyme. Subsequently, allatom molecular dynamics (MD) simulations and binding free energy calculations were employed to elucidate the structural insights into the protein-ligand complexations. RESULTS: Rosmarinic acid showed the highest inhibitory activity against NA with the IC50 of 0.40 µM compared to caffeic acid (IC50 of 0.81 µM) and salvianolic acid A (IC50 of >1 µM). From 100-ns MD simulations, the binding affinity, hot-spot residues, and H-bond formations of rosmarinic acid/NA complex were higher than those of caffeic acid/NA model, in which their molecular complexations was driven mainly by electrostatic attractions and H-bond formations from several charged residues (R118, E119, D151, R152, E227, E277, and R371). Notably, the two hydroxyl groups on both phenyl and phenylacetic rings of rosmarinic acid play a crucial role in stabilizing NA through a strongly formed Hbond( s). CONCLUSION: Our findings shed light on the potentiality of rosmarinic acid as a lead compound for further development of a potential influenza NA inhibitor.


Asunto(s)
Antivirales/farmacología , Cinamatos/farmacología , Depsidos/farmacología , Inhibidores Enzimáticos/farmacología , Gripe Humana/tratamiento farmacológico , Simulación de Dinámica Molecular , Neuraminidasa/antagonistas & inhibidores , Antivirales/química , Ácidos Cafeicos/química , Ácidos Cafeicos/farmacología , Cinamatos/química , Depsidos/química , Inhibidores Enzimáticos/química , Humanos , Gripe Humana/enzimología , Ligandos , Estructura Molecular , Neuraminidasa/metabolismo , Termodinámica , Ácido Rosmarínico
3.
J Virol ; 93(21)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31391268

RESUMEN

Cleavage of influenza virus hemagglutinin (HA) by host cell proteases is essential for virus infectivity and spread. We previously demonstrated in vitro that the transmembrane protease TMPRSS2 cleaves influenza A virus (IAV) and influenza B virus (IBV) HA possessing a monobasic cleavage site. Subsequent studies revealed that TMPRSS2 is crucial for the activation and pathogenesis of H1N1pdm and H7N9 IAV in mice. In contrast, activation of H3N2 IAV and IBV was found to be independent of TMPRSS2 expression and supported by an as-yet-undetermined protease(s). Here, we investigated the role of TMPRSS2 in proteolytic activation of IAV and IBV in three human airway cell culture systems: primary human bronchial epithelial cells (HBEC), primary type II alveolar epithelial cells (AECII), and Calu-3 cells. Knockdown of TMPRSS2 expression was performed using a previously described antisense peptide-conjugated phosphorodiamidate morpholino oligomer, T-ex5, that interferes with splicing of TMPRSS2 pre-mRNA, resulting in the expression of enzymatically inactive TMPRSS2. T-ex5 treatment produced efficient knockdown of active TMPRSS2 in all three airway cell culture models and prevented proteolytic activation and multiplication of H7N9 IAV in Calu-3 cells and H1N1pdm, H7N9, and H3N2 IAV in HBEC and AECII. T-ex5 treatment also inhibited the activation and spread of IBV in AECII but did not affect IBV activation in HBEC and Calu-3 cells. This study identifies TMPRSS2 as the major HA-activating protease of IAV in human airway cells and IBV in type II pneumocytes and as a potential target for the development of novel drugs to treat influenza infections.IMPORTANCE Influenza A viruses (IAV) and influenza B viruses (IBV) cause significant morbidity and mortality during seasonal outbreaks. Cleavage of the viral surface glycoprotein hemagglutinin (HA) by host proteases is a prerequisite for membrane fusion and essential for virus infectivity. Inhibition of relevant proteases provides a promising therapeutic approach that may avoid the development of drug resistance. HA of most influenza viruses is cleaved at a monobasic cleavage site, and a number of proteases have been shown to cleave HA in vitro This study demonstrates that the transmembrane protease TMPRSS2 is the major HA-activating protease of IAV in primary human bronchial cells and of both IAV and IBV in primary human type II pneumocytes. It further reveals that human and murine airway cells can differ in their HA-cleaving protease repertoires. Our data will help drive the development of potent and selective protease inhibitors as novel drugs for influenza treatment.


Asunto(s)
Virus de la Influenza A/fisiología , Virus de la Influenza B/fisiología , Gripe Humana/virología , Serina Endopeptidasas/metabolismo , Animales , Bronquios/citología , Células Cultivadas , Células Epiteliales/virología , Técnicas de Silenciamiento del Gen , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Interacciones Huésped-Patógeno , Humanos , Gripe Humana/enzimología , Gripe Humana/metabolismo , Ratones , Infecciones por Orthomyxoviridae/enzimología , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Alveolos Pulmonares/citología , Serina Endopeptidasas/genética , Regulación hacia Arriba , Replicación Viral
4.
Viruses ; 11(2)2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30791550

RESUMEN

Influenza A viruses (IAVs) are a major cause of respiratory illness and are responsible for yearly epidemics associated with more than 500,000 annual deaths globally. Novel IAVs may cause pandemic outbreaks and zoonotic infections with, for example, highly pathogenic avian influenza virus (HPAIV) of the H5N1 and H7N9 subtypes, which pose a threat to public health. Treatment options are limited and emergence of strains resistant to antiviral drugs jeopardize this even further. Like all viruses, IAVs depend on host factors for every step of the virus replication cycle. Host kinases link multiple signaling pathways in respond to a myriad of stimuli, including viral infections. Their regulation of multiple response networks has justified actively targeting cellular kinases for anti-cancer therapies and immune modulators for decades. There is a growing volume of research highlighting the significant role of cellular kinases in regulating IAV infections. Their functional role is illustrated by the required phosphorylation of several IAV proteins necessary for replication and/or evasion/suppression of the innate immune response. Identified in the majority of host factor screens, functional studies further support the important role of kinases and their potential as host restriction factors. PKC, ERK, PI3K and FAK, to name a few, are kinases that regulate viral entry and replication. Additionally, kinases such as IKK, JNK and p38 MAPK are essential in mediating viral sensor signaling cascades that regulate expression of antiviral chemokines and cytokines. The feasibility of targeting kinases is steadily moving from bench to clinic and already-approved cancer drugs could potentially be repurposed for treatments of severe IAV infections. In this review, we will focus on the contribution of cellular kinases to IAV infections and their value as potential therapeutic targets.


Asunto(s)
Interacciones Huésped-Patógeno , Gripe Humana/enzimología , Fosfotransferasas/metabolismo , Internalización del Virus , Antivirales/uso terapéutico , Línea Celular , Humanos , Quinasa I-kappa B/metabolismo , Inmunidad Innata , Subtipo H1N1 del Virus de la Influenza A , Subtipo H5N1 del Virus de la Influenza A , Subtipo H7N9 del Virus de la Influenza A , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Postepy Biochem ; 63(4): 261-268, 2017.
Artículo en Polaco | MEDLINE | ID: mdl-29374427

RESUMEN

From the earliest times, medicine has focused on finding the most suitable and effective treatment for every patient. At present, a dynamic development of diagnostic methods and techniques for designing new drugs allows to create therapies for many diseases at the molecular level. Among the many drugs appearing on the medical market every year, special attention should be paid to those whose action is based on the inhibition of proteolytic enzyme activity. Protease inhibitors are a diverse group of biologically active molecules for which antiviral, antimicrobial, antifungal, antiparasitic or anticancer effects have been documented. Successes in the treatment of HIV infection, hepatitis C and influenza diseases certainly encourage researchers to look for new inhibitors that could be used in new therapies. This paper provides an overview of selected information on enzyme inhibitors, especially protease inhibitors, which are already registered medicines and substances that are promising candidates for medical use.


Asunto(s)
Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/uso terapéutico , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antivirales/farmacología , Antivirales/uso terapéutico , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/enzimología , Hepatitis C/tratamiento farmacológico , Hepatitis C/enzimología , Humanos , Gripe Humana/tratamiento farmacológico , Gripe Humana/enzimología
6.
Am J Physiol Lung Cell Mol Physiol ; 310(7): L689-99, 2016 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-26747784

RESUMEN

Influenza A viruses (IAV) trigger contagious acute respiratory diseases. A better understanding of the molecular mechanisms of IAV pathogenesis and host immune responses is required for the development of more efficient treatments of severe influenza. Calpains are intracellular proteases that participate in diverse cellular responses, including inflammation. Here, we used in vitro and in vivo approaches to investigate the role of calpain signaling in IAV pathogenesis. Calpain expression and activity were found altered in IAV-infected bronchial epithelial cells. With the use of small-interfering RNA (siRNA) gene silencing, specific synthetic inhibitors of calpains, and mice overexpressing calpastatin, we found that calpain inhibition dampens IAV replication and IAV-triggered secretion of proinflammatory mediators and leukocyte infiltration. Remarkably, calpain inhibition has a protective impact in IAV infection, since it significantly reduced mortality of mice challenged not only by seasonal H3N2- but also by hypervirulent H5N1 IAV strains. Hence, our study suggests that calpains are promising therapeutic targets for treating IAV acute pneumonia.


Asunto(s)
Calpaína/fisiología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Gripe Humana/enzimología , Animales , Citocinas/metabolismo , Perros , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Viral de la Expresión Génica , Humanos , Gripe Humana/inmunología , Gripe Humana/virología , Células de Riñón Canino Madin Darby , Ratones Endogámicos C57BL , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal , Replicación Viral
7.
Viruses ; 8(1)2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26761025

RESUMEN

Highly pathogenic H5N1 infections are often accompanied by excessive pro-inflammatory response, high viral titer, and apoptosis; as such, the efficient control of these infections poses a great challenge. The pathogenesis of influenza virus infection is also related to oxidative stress. However, the role of endogenic genes with antioxidant effect in the control of influenza viruses, especially H5N1 viruses, should be further investigated. In this study, the H5N1 infection in lung epithelial cells decreased Cu/Zn superoxide dismutase (SOD1) expression at mRNA and protein levels. Forced SOD1 expression significantly inhibited the H5N1-induced increase in reactive oxygen species, decreased pro-inflammatory response, prevented p65 and p38 phosphorylation, and impeded viral ribonucleoprotein nuclear export and viral replication. The SOD1 overexpression also rescued H5N1-induced cellular apoptosis and alleviated H5N1-caused mitochondrial dysfunction. Therefore, this study described the role of SOD1 in the replication of H5N1 influenza virus and emphasized the relevance of this enzyme in the control of H5N1 replication in epithelial cells. Pharmacological modulation or targeting SOD1 may open a new way to fight H5N1 influenza virus.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/fisiología , Gripe Humana/enzimología , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Replicación Viral , Muerte Celular , Línea Celular , Células Epiteliales/citología , Células Epiteliales/enzimología , Células Epiteliales/metabolismo , Células Epiteliales/virología , Interacciones Huésped-Patógeno , Humanos , Subtipo H5N1 del Virus de la Influenza A/genética , Gripe Humana/genética , Gripe Humana/fisiopatología , Gripe Humana/virología , Pulmón/citología , Pulmón/enzimología , Pulmón/metabolismo , Pulmón/virología , Superóxido Dismutasa/genética , Superóxido Dismutasa-1
8.
J Virol ; 88(18): 10432-47, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24942585

RESUMEN

UNLABELLED: The influenza A virus genome possesses eight negative-strand RNA segments in the form of viral ribonucleoprotein particles (vRNPs) in association with the three viral RNA polymerase subunits (PB2, PB1, and PA) and the nucleoprotein (NP). Through interactions with multiple host factors, the RNP subunits play vital roles in replication, host adaptation, interspecies transmission, and pathogenicity. In order to gain insight into the potential roles of RNP subunits in the modulation of the host's innate immune response, the interactions of each RNP subunit with retinoic acid-inducible gene I protein (RIG-I) from mammalian and avian species were investigated. Studies using coimmunoprecipitation (co-IP), bimolecular fluorescence complementation (BiFc), and colocalization using confocal microscopy provided direct evidence for the RNA-independent binding of PB2, PB1, and PA with RIG-I from various hosts (human, swine, mouse, and duck). In contrast, the binding of NP with RIG-I was found to be RNA dependent. Expression of the viral NS1 protein, which interacts with RIG-I, did not interfere with the association of RNA polymerase subunits with RIG-I. The association of each individual virus polymerase component with RIG-I failed to significantly affect the interferon (IFN) induction elicited by RIG-I and 5' triphosphate (5'ppp) RNA in reporter assays, quantitative reverse transcription-PCR (RT-PCR), and IRF3 phosphorylation tests. Taken together, these findings indicate that viral RNA polymerase components PB2, PB1, and PA directly target RIG-I, but the exact biological significance of these interactions in the replication and pathogenicity of influenza A virus needs to be further clarified. IMPORTANCE: RIG-I is an important RNA sensor to elicit the innate immune response in mammals and some bird species (such as duck) upon influenza A virus infection. Although the 5'-triphosphate double-stranded RNA (dsRNA) panhandle structure at the end of viral genome RNA is responsible for the binding and subsequent activation of RIG-I, this structure is supposedly wrapped by RNA polymerase complex (PB2, PB1, and PA), which may interfere with the induction of RIG-I signaling pathway. In the present study, PB2, PB1, and PA were found to individually interact with RIG-Is from multiple mammalian and avian species in an RNA-independent manner, without significantly affecting the generation of IFN. The data suggest that although RIG-I binding by RNA polymerase complex is conserved in different species, it does not appear to play crucial role in the modulation of IFN in vitro.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Subtipo H9N2 del Virus de la Influenza A/enzimología , Gripe Aviar/enzimología , Gripe Humana/enzimología , ARN Polimerasa Dependiente del ARN/metabolismo , Enfermedades de los Porcinos/enzimología , Proteínas Virales/metabolismo , Animales , Línea Celular , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , Patos , Humanos , Subtipo H9N2 del Virus de la Influenza A/genética , Gripe Aviar/genética , Gripe Aviar/virología , Gripe Humana/genética , Gripe Humana/virología , Ratones , Infecciones por Orthomyxoviridae/enzimología , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/veterinaria , Infecciones por Orthomyxoviridae/virología , Unión Proteica , ARN Polimerasa Dependiente del ARN/genética , Receptores Inmunológicos , Porcinos , Enfermedades de los Porcinos/genética , Proteínas Virales/genética
9.
J Biomol Screen ; 19(6): 936-46, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24464431

RESUMEN

During viral infection of human cells, host kinases mediate signaling activities that are used by all viruses for replication; therefore, targeting of host kinases is of broad therapeutic interest. Here, host kinases were globally screened during human influenza virus (H1N1) infection to determine the time-dependent effects of virus infection and replication on kinase function. Desthiobiotin-labeled analogs of adenosine triphosphate and adenosine diphosphate were used to probe and covalently label host kinases in infected cell lysates, and probe affinity was determined. Using infected human A549 cells, we screened for time-dependent signal changes and identified host kinases whose probe affinities differed significantly when compared to uninfected cells. Our screen identified 10 novel host kinases that have not been previously shown to be involved with influenza virus replication, and we validated the functional importance of these novel kinases during infection using targeted small interfering RNAs (siRNAs). The effects of kinase-targeted siRNA knockdowns on replicating virus levels were measured by quantitative reverse-transcription PCR and cytoprotection assays. We identified several novel host kinases that, when knocked down, enhanced or reduced the viral load in cell culture. This preliminary work represents the first screen of the changing host kinome in influenza virus-infected human cells.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/enzimología , Proteínas Serina-Treonina Quinasas/química , ARN Interferente Pequeño/genética , Replicación Viral , Células A549 , Adenosina Difosfato/química , Adenosina Trifosfato/química , Apoptosis , Biotina/análogos & derivados , Biotina/química , Supervivencia Celular , Cromatografía Liquida , Descubrimiento de Drogas , Humanos , Espectrometría de Masas , Quinasa 1 Relacionada con NIMA/química , Péptidos/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina-Treonina Quinasa 3 , Carga Viral
10.
Hong Kong Med J ; 19 Suppl 4: 29-35, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23775184

RESUMEN

1. Cyclooxygenase-2 (COX-2), along with TNF-α and other proinflammatory cytokines, was hyperinduced in H5N1- infected macrophages in vitro and in epithelial cells of autopsied lung tissues of infected patients. 2. The COX-2 mediated amplification of the proinflammatory response is rapid, and the effects elicited by the H5N1-triggered proinflammatory cascade are broader than those arising from direct viral infection. 3. Selective COX-2 inhibitors suppress the H5N1- hyperinduced cytokines in the proinflammatory cascade.


Asunto(s)
Antivirales/farmacología , Inhibidores de la Ciclooxigenasa 2/farmacología , Subtipo H5N1 del Virus de la Influenza A/enzimología , Gripe Humana/enzimología , Línea Celular Tumoral , Ciclooxigenasa 2/efectos de los fármacos , Citocinas/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/virología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/virología , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Gripe Humana/tratamiento farmacológico , Gripe Humana/virología , Pulmón/citología , Pulmón/virología , Macrófagos/metabolismo , Macrófagos/virología , Factor de Necrosis Tumoral alfa/metabolismo
11.
PLoS One ; 8(6): e66796, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23805279

RESUMEN

Human protein kinases (HPKs) have profound effects on cellular responses. To better understand the role of HPKs and the signaling networks that influence influenza virus replication, a small interfering RNA (siRNA) screen of 720 HPKs was performed. From the screen, 17 HPKs (NPR2, MAP3K1, DYRK3, EPHA6, TPK1, PDK2, EXOSC10, NEK8, PLK4, SGK3, NEK3, PANK4, ITPKB, CDC2L5 (CDK13), CALM2, PKN3, and HK2) were validated as essential for A/WSN/33 influenza virus replication, and 6 HPKs (CDK13, HK2, NEK8, PANK4, PLK4 and SGK3) were identified as vital for both A/WSN/33 and A/New Caledonia/20/99 influenza virus replication. These HPKs were found to affect multiple host pathways and regulated by miRNAs induced during infection. Using a panel of miRNA agonists and antagonists, miR-149* was found to regulate NEK8 expression, miR-548d-3p was found to regulate MAPK1 transcript expression, and miRs -1228 and -138 to regulate CDK13 expression. Up-regulation of miR-34c induced PLK4 transcript and protein expression and enhanced influenza virus replication, while miR-34c inhibition reduced viral replication. These findings identify HPKs important for influenza viral replication and show the miRNAs that govern their expression.


Asunto(s)
Virus de la Influenza A/fisiología , Gripe Humana/genética , Gripe Humana/virología , MicroARNs/metabolismo , Proteínas Quinasas/metabolismo , Replicación Viral/genética , Células A549 , Animales , Secuencia de Bases , Perros , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/aislamiento & purificación , Gripe Humana/enzimología , Gripe Humana/patología , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Quinasa 1 de Quinasa de Quinasa MAP/genética , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Células de Riñón Canino Madin Darby , MicroARNs/agonistas , MicroARNs/antagonistas & inhibidores , Quinasas Relacionadas con NIMA/antagonistas & inhibidores , Quinasas Relacionadas con NIMA/genética , Quinasas Relacionadas con NIMA/metabolismo , Proteínas de la Nucleocápside , Fenotipo , Proteínas Quinasas/química , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Ribosómico 18S/genética , ARN Ribosómico 18S/metabolismo , Proteínas de Unión al ARN/metabolismo , Alineación de Secuencia , Regulación hacia Arriba , Proteínas del Núcleo Viral/metabolismo
12.
PLoS One ; 8(6): e67616, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23799150

RESUMEN

The 2009/2010 pandemic influenza virus (H1N1pdm) contains an avian-lineage PB2 gene that lacks E627K and D701N substitutions important in the pathogenesis and transmission of avian-origin viruses in humans or other mammals. Previous studies have shown that PB2-627K is not necessary because of a compensatory Q591R substitution. The role that PB2-701N plays in the H1N1pdm phenotype is not well understood. Therefore, PB2-D701N was introduced into an H1N1pdm virus (A/New York/1682/2009 (NY1682)) and analyzed in vitro and in vivo. Mini-genome replication assay, in vitro replication characteristics in cell lines, and analysis in the mouse and ferret models demonstrated that PB2-D701N increased virus replication rates and resulted in more severe pathogenicity in mice and more efficient transmission in ferrets. In addition, compared to the NY1682-WT virus, the NY1682-D701N mutant virus induced less IFN-λ and replicated to a higher titer in primary human alveolar epithelial cells. These findings suggest that the acquisition of the PB2-701N substitution by H1N1pdm viruses may result in more severe disease or increase transmission in humans.


Asunto(s)
Sustitución de Aminoácidos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/transmisión , ARN Polimerasa Dependiente del ARN/genética , Proteínas Virales/genética , Replicación Viral/genética , Células Epiteliales Alveolares/inmunología , Células Epiteliales Alveolares/virología , Animales , Asparagina/genética , Línea Celular Tumoral , Citocinas/metabolismo , Perros , Femenino , Hurones , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Gripe Humana/enzimología , Gripe Humana/virología , Cinética , Masculino , Ratones , Ratones Endogámicos BALB C
13.
Curr Microbiol ; 67(4): 431-6, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23680974

RESUMEN

Patchouli alcohol (PA) is a kind of methanol extracted from traditional Chinese medicine Pogostemonis Herba. Our research aimed to observe the anti-influenza virus role of PA in vitro. 16HBE (human respiratory epithelial cell) was infected by H1N1 (A/FM1/1/47) to set the cell model. Then the 16HBE was co-cultivated with three kinds of immune cells: dendritic cells, macrophages, and monocytes, PA (the concentration is 10 µg/mL) was added as a treatment intervention for 24 h. The immune cells and the supernate were collected for RT-PCR and ELISA detection related to RLH (RIG-1-like helicases) pathway. Results showed that the IL-4 and IFN-γ in supernate were increased after H1N1 infection, and the PA treatment suppressed the expression of cytokines and the mRNA of RLH pathway. PA anti-influenza virus may through regulate the RLH singal pathway.


Asunto(s)
Antivirales/farmacología , Medicamentos Herbarios Chinos/farmacología , Factores Inmunológicos/farmacología , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Gripe Humana/inmunología , ARN Helicasas/inmunología , Sesquiterpenos/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Humanos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/tratamiento farmacológico , Gripe Humana/enzimología , Gripe Humana/virología , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , ARN Helicasas/genética , Transducción de Señal/efectos de los fármacos
14.
Ter Arkh ; 85(3): 58-62, 2013.
Artículo en Ruso | MEDLINE | ID: mdl-23720844

RESUMEN

AIM: To study the specific features of cytokine gene polymorphisms (such as TNF G308A, IL10 C592A, IL10 C819T, IL10 G1082A) and vascular tone regulatory gene polymorphism (eNOS C786T) in patients with influenza A/H1N1 complicated by pneumonia. SUBJECTS AND METHODS: Patients treated for pneumonia in the presence of influenza A/H1N1, divided into 3 groups: 1) 37 patients with severe pneumonia; 2) 74 with mild pneumonia; and 3) 68 healthy individuals, were examined. Molecular genetic testing was made using a polymerase chain reaction technique. Serum TNFalpha, IL-10 and s-ICAM-1 concentrations were measured by enzyme immunoassay. RESULTS: As compared to the control group, the patients with influenza-related pneumonia were more frequently homozygous for the G allele of the TNF 308 G/A polymorphism. The IL-10 G allele (1082 G/A) was considerably prevalent as homozygous carriage and the IL-10 G allele (592 G/A) was as a homozygous type to a greater extent. An abnormal zygote of the eNOS T/T (786 C/T) polymorphism was found to be prevalent among the patients with influenza-related pneumonia; on the contrary, there were mainly heterozygous C/T carriers in the control group. In the C/T genotype group, the level of soluble intercellular adhesion molecule 1 (sICAM-1) was lower than in other polymorphic variants (786 C/T). CONCLUSION; The study of the patients' genetic status in influenza A/H1N1 will be able to evaluate the severity of the disease and to predict possible complications.


Asunto(s)
Gripe Humana/genética , Interleucina-10/genética , Óxido Nítrico Sintasa de Tipo III/genética , Neumonía Viral/genética , Polimorfismo Genético/genética , Factor de Necrosis Tumoral alfa/genética , Adulto , Alelos , Femenino , Tamización de Portadores Genéticos , Predisposición Genética a la Enfermedad , Homocigoto , Humanos , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Gripe Humana/enzimología , Gripe Humana/inmunología , Masculino , Persona de Mediana Edad , Neumonía Viral/etiología , Neumonía Viral/inmunología
15.
Int J Mol Sci ; 14(4): 7327-40, 2013 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-23549267

RESUMEN

Influenza viruses of avian origin continue to pose pandemic threats to human health. Some of the H5N1 and H9N2 virus subtypes induce markedly elevated cytokine levels when compared with the seasonal H1N1 virus. We previously showed that H5N1/97 hyperinduces tumor necrosis factor (TNF)-alpha through p38 mitogen activated protein kinase (MAPK). However, the detailed mechanisms of p38MAPK activation and TNF-alpha hyperinduction following influenza virus infections are not known. Negative feedback regulations of cytokine expression play important roles in avoiding overwhelming production of proinflammatory cytokines. Here we hypothesize that protein phosphatases are involved in the regulation of cytokine expressions during influenza virus infection. We investigated the roles of protein phosphatases including MAPK phosphatase-1 (MKP-1) and protein phosphatase type 2A (PP2A) in modulating p38MAPK activation and downstream TNF-alpha expressions in primary human monocyte-derived macrophages (PBMac) infected with H9N2/G1 or H1N1 influenza virus. We demonstrate that H9N2/G1 virus activated p38MAPK and hyperinduced TNF-alpha production in PBMac when compared with H1N1 virus. H9N2/G1 induced PP2A activity in PBMac and, with the treatment of a PP2A inhibitor, p38MAPK phosphorylation and TNF-alpha production were further increased in the virus-infected macrophages. However, H9N2/G1 did not induce the expression of PP2A indicating that the activation of PP2A is not mediated by p38MAPK in virus-infected PBMac. On the other hand, PP2A may not be the targets of H9N2/G1 in the upstream of p38MAPK signaling pathways since H1N1 also induced PP2A activation in primary macrophages. Our results may provide new insights into the control of cytokine dysregulation.


Asunto(s)
Gripe Humana/enzimología , Gripe Humana/patología , Proteína Fosfatasa 2/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Dominio Catalítico , Células Cultivadas , Fosfatasa 1 de Especificidad Dual/metabolismo , Activación Enzimática , Humanos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H9N2 del Virus de la Influenza A/fisiología , Gripe Humana/sangre , Gripe Humana/virología , Macrófagos/enzimología , Macrófagos/patología , Fosforilación
16.
J Virol ; 87(9): 4861-71, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23408626

RESUMEN

The influenza A virus NS1 protein affects virulence through several mechanisms, including the host's innate immune response and various signaling pathways. Highly pathogenic avian influenza (HPAI) viruses of the H5N1 subtype continue to evolve through reassortment and mutations. Our recent phylogenetic analysis identified a group of HPAI H5N1 viruses with two characteristic mutations in NS1: the avian virus-type PDZ domain-binding motif ESEV (which affects virulence) was replaced with ESKV, and NS1-138F (which is highly conserved among all influenza A viruses and may affect the activation of the phosphatidylinositol 3-kinase [PI3K]/Akt signaling pathway) was replaced with NS1-138Y. Here, we show that an HPAI H5N1 virus (A/duck/Hunan/69/2004) encoding NS1-ESKV and NS1-138Y was confined to the respiratory tract of infected mice, whereas a mutant encoding NS1-ESEV and NS1-138F caused systemic infection and killed mice more efficiently. Mutation of either one of these sites had small effects on virulence. In addition, we found that the amino acid at NS1-138 affected not only the induction of the PI3K/Akt pathway but also the interaction of NS1 with cellular PDZ domain proteins. Similarly, the mutation in the PDZ domain-binding motif of NS1 altered its binding to cellular PDZ domain proteins and affected Akt phosphorylation. These findings suggest a functional interplay between the mutations at NS1-138 and NS1-229 that results in a synergistic effect on influenza virulence.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/metabolismo , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Gripe Humana/virología , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Animales , Línea Celular , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Femenino , Células HEK293 , Humanos , Subtipo H5N1 del Virus de la Influenza A/química , Subtipo H5N1 del Virus de la Influenza A/genética , Gripe Humana/enzimología , Gripe Humana/genética , Ratones , Ratones Endogámicos BALB C , Dominios PDZ , Unión Proteica , Estructura Terciaria de Proteína , Proteínas no Estructurales Virales/genética , Virulencia
17.
Viruses ; 5(1): 182-91, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23322171

RESUMEN

Cyclophilin A (CypA) is the main member of the immunophilin superfamily that has peptidyl-prolyl cis-trans isomerase activity. CypA participates in protein folding, cell signaling, inflammation and tumorigenesis. Further, CypA plays critical roles in the replication of several viruses. Upon influenza virus infection, CypA inhibits viral replication by interacting with the M1 protein. In addition, CypA is incorporated into the influenza virus virions. Finally, Cyclosporin A (CsA), the main inhibitor of CypA, inhibits influenza virus replication through CypA-dependent and -independent pathways. This review briefly summarizes recent advances in understanding the roles of CypA during influenza virus infection.


Asunto(s)
Ciclofilina A/metabolismo , Gripe Humana/enzimología , Gripe Humana/virología , Orthomyxoviridae/fisiología , Animales , Ciclofilina A/genética , Humanos , Gripe Humana/genética , Orthomyxoviridae/genética , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo , Replicación Viral
18.
FASEB J ; 26(7): 2911-8, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22490782

RESUMEN

Underlying mechanisms of individual variation in severity of influenza infection and response to vaccination are poorly understood. We investigated the effect of reduced heme oxygenase-1 (HO-1) expression on vaccine response and outcome of influenza infection. HO-1-deficient and wild-type (WT) mice (kingdom, Animalia; phylum, Chordata; genus/species, Mus musculus) were infected with influenza virus A/PR/8/34 with or without prior vaccination with an adenoviral-based influenza vaccine. A genome-wide association study evaluated the expression of single-nucleotide polymorphisms (SNPs) in the HO-1 gene and the response to influenza vaccination in healthy humans. HO-1-deficient mice had decreased survival after influenza infection compared to WT mice (median survival 5.5 vs. 6.5 d, P=0.016). HO-1-deficient mice had impaired production of antibody following influenza vaccination compared to WT mice (mean antibody titer 869 vs. 1698, P=0.02). One SNP in HO-1 and one SNP in the constitutively expressed isoform HO-2 were independently associated with decreased antibody production after influenza vaccination in healthy human volunteers (P=0.017 and 0.014, respectively). HO-1 deficient mice were paired with sex- and age-matched WT controls. HO-1 affects the immune response to both influenza infection and vaccination, suggesting that therapeutic induction of HO-1 expression may represent a novel adjuvant to enhance influenza vaccine effectiveness.


Asunto(s)
Envejecimiento/inmunología , Envejecimiento/metabolismo , Hemo-Oxigenasa 1/metabolismo , Proteínas de la Membrana/metabolismo , Infecciones por Orthomyxoviridae/enzimología , Infecciones por Orthomyxoviridae/inmunología , Animales , Anticuerpos Antivirales/biosíntesis , Secuencia de Bases , Cartilla de ADN/genética , Femenino , Estudio de Asociación del Genoma Completo , Hemo-Oxigenasa 1/deficiencia , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/inmunología , Humanos , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/enzimología , Gripe Humana/inmunología , Gripe Humana/prevención & control , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/virología , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Vacunación
19.
PLoS One ; 7(3): e33732, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22470468

RESUMEN

The host response to the low pathogenic avian influenza (LPAI) H5N2, H5N3 and H9N2 viruses were examined in A549, MDCK, and CEF cells using a systems-based approach. The H5N2 and H5N3 viruses replicated efficiently in A549 and MDCK cells, while the H9N2 virus replicated least efficiently in these cell types. However, all LPAI viruses exhibited similar and higher replication efficiencies in CEF cells. A comparison of the host responses of these viruses and the H1N1/WSN virus and low passage pH1N1 clinical isolates was performed in A549 cells. The H9N2 and H5N2 virus subtypes exhibited a robust induction of Type I and Type III interferon (IFN) expression, sustained STAT1 activation from between 3 and 6 hpi, which correlated with large increases in IFN-stimulated gene (ISG) expression by 10 hpi. In contrast, cells infected with the pH1N1 or H1N1/WSN virus showed only small increases in Type III IFN signalling, low levels of ISG expression, and down-regulated expression of the IFN type I receptor. JNK activation and increased expression of the pro-apoptotic XAF1 protein was observed in A549 cells infected with all viruses except the H1N1/WSN virus, while MAPK p38 activation was only observed in cells infected with the pH1N1 and the H5 virus subtypes. No IFN expression and low ISG expression levels were generally observed in CEF cells infected with either AIV, while increased IFN and ISG expression was observed in response to the H1N1/WSN infection. These data suggest differences in the replication characteristics and antivirus signalling responses both among the different LPAI viruses, and between these viruses and the H1N1 viruses examined. These virus-specific differences in host cell signalling highlight the importance of examining the host response to avian influenza viruses that have not been extensively adapted to mammalian tissue culture.


Asunto(s)
Células Epiteliales/metabolismo , Gripe Humana/patología , Interferón Tipo I/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Reguladoras de la Apoptosis , Aves , Línea Celular Tumoral , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H5N2 del Virus de la Influenza A/genética , Subtipo H5N2 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H9N2 del Virus de la Influenza A/genética , Subtipo H9N2 del Virus de la Influenza A/crecimiento & desarrollo , Gripe Aviar/genética , Gripe Aviar/virología , Gripe Humana/enzimología , Interferón Tipo I/genética , Interferones , Interleucinas/genética , Interleucinas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas de Neoplasias/metabolismo , ARN Viral/metabolismo , Receptor de Interferón alfa y beta/metabolismo , Factor de Transcripción STAT1/metabolismo , Replicación Viral , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
J Biol Chem ; 287(15): 11899-910, 2012 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-22343630

RESUMEN

We previously reported that IL-27, which belongs to the IL-12 family of cytokines, is elevated in the serum of patients infected with influenza A virus (IAV). Here, we show that the expression of IL-27 was significantly up-regulated in A549 human lung epithelial cells and human peripheral blood mononuclear cells infected with IAV. Additionally, IAV triggered IL-27 expression through protein kinase A and cAMP-response element-binding protein signaling, which was mediated by cyclooxygenase-2-derived prostaglandin E(2). IL-27 inhibited IAV replication by STAT1/2/3 phosphorylation and activated antiviral factor protein kinase R phosphorylation. Clinical analysis showed that IL-27 levels were significantly elevated in a cohort of patients infected with IAV compared with healthy individuals and that circulating IL-27 levels were tightly and positively correlated with prostaglandin E(2) levels. These results indicate that IL-27 expression is one host immune factor produced in response to IAV infection and that elevated IL-27 levels inhibit viral replication.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Ciclooxigenasa 2/metabolismo , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H3N2 del Virus de la Influenza A/fisiología , Gripe Humana/enzimología , Interleucinas/metabolismo , Animales , Sitios de Unión , Estudios de Casos y Controles , Línea Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Dinoprostona/sangre , Perros , Activación Enzimática , Humanos , Gripe Humana/sangre , Gripe Humana/virología , Interleucinas/sangre , Interleucinas/genética , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/virología , Antígenos de Histocompatibilidad Menor , Fosforilación , Regiones Promotoras Genéticas , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Activación Transcripcional , Replicación Viral , eIF-2 Quinasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA