Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 166
Filtrar
1.
J Virol ; 97(10): e0060223, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37754760

RESUMO

IMPORTANCE: Influenza A viruses (IAVs) contain hemagglutinin (HA) proteins involved in sialoglycan receptor binding and neuraminidase (NA) proteins that cleave sialic acids. While the importance of the NA protein in virion egress is well established, its role in virus entry remains to be fully elucidated. NA activity is needed for the release of virions from mucus decoy receptors, but conflicting results have been reported on the importance of NA activity in virus entry in the absence of decoy receptors. We now show that inhibition of NA activity affects virus entry depending on the receptor-binding properties of HA and the receptor repertoire present on cells. Inhibition of entry by the presence of mucus correlated with the importance of NA activity for virus entry, with the strongest inhibition being observed when mucus and OsC were combined. These results shed light on the importance in virus entry of the NA protein, an important antiviral drug target.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza , Vírus da Influenza A , Neuraminidase , Receptores Virais , Proteínas Virais , Internalização do Vírus , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Vírus da Influenza A/enzimologia , Vírus da Influenza A/metabolismo , Influenza Humana/enzimologia , Influenza Humana/metabolismo , Neuraminidase/antagonistas & inibidores , Neuraminidase/metabolismo , Ligação Proteica , Receptores Virais/metabolismo , Especificidade por Substrato , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo , Linhagem Celular , Muco
2.
PLoS Pathog ; 18(9): e1010794, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36070294

RESUMO

Influenza virus has the ability to circumvent host innate immune system through regulating certain host factors for its effective propagation. However, the detailed mechanism is still not fully understood. Here, we report that a host sphingolipid metabolism-related factor, sphingosine kinase 2 (SPHK2), upregulated during influenza A virus (IAV) infection, promotes IAV infection in an enzymatic independent manner. The enhancement of the virus replication is not abolished in the catalytic-incompetent SPHK2 (G212E) overexpressing cells. Intriguingly, the sphingosine-1-phosphate (S1P) related factor HDAC1 also plays a crucial role in SPHK2-mediated IAV infection. We found that SPHK2 cannot facilitate IAV infection in HDAC1 deficient cells. More importantly, SPHK2 overexpression diminishes the IFN-ß promoter activity upon IAV infection, resulting in the suppression of type I IFN signaling. Furthermore, ChIP-qPCR assay revealed that SPHK2 interacts with IFN-ß promoter through the binding of demethylase TET3, but not with the other promoters regulated by TET3, such as TGF-ß1 and IL6 promoters. The specific regulation of SPHK2 on IFN-ß promoter through TET3 can in turn recruit HDAC1 to the IFN-ß promoter, enhancing the deacetylation of IFN-ß promoter, therefore leading to the inhibition of IFN-ß transcription. These findings reveal an enzymatic independent mechanism on host SPHK2, which associates with TET3 and HDAC1 to negatively regulate type I IFN expression and thus facilitates IAV propagation.


Assuntos
Influenza Humana , Interferon beta , Fosfotransferases (Aceptor do Grupo Álcool) , Humanos , Vírus da Influenza A , Influenza Humana/enzimologia , Interferon beta/genética , Interferon beta/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Replicação Viral
3.
Viruses ; 14(2)2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-35215907

RESUMO

The identification and elucidation of host pathways for viral infection are critical for understanding the viral infection processes and novel therapeutics development. Here, for the first time, we discover that the human SUMOylation pathway is essential for the IBV viral life cycle. First, IBV viruses were completely inhibited by a novel SUMOylation specific inhibitor, STE025, discovered from our FRET-based high-throughput screening, and the inhibition was very potent, with IC50~ 0.1 µM in an IBV-induced cell death rescue assay; Second, we determined that the IBV M1 protein was SUMOylated, which was mediated by the SUMOylation E2 conjugation enzyme and the E3 ligase enzyme at very high affinities, of 0.20 µM and 0.22 µM, respectively; Third, the mutation of the IBV M1 SUMOylation site, K21R, completely abolished the viral particle generation, strongly suggesting the requirement of SUMOylation for the IBV life cycle. These results suggest that the blockage of the host human SUMOylation pathway is very effective for IBV inhibition. We therefore propose that the host SUMOylation pathway is a critical host factor for the IBV virus life cycle. The identification and inhibition of critical host factor(s) provide a novel strategy for future anti-viral therapeutics development, such as IBV and other viruses.


Assuntos
Vírus da Influenza B/metabolismo , Influenza Humana/virologia , Sumoilação , Proteínas da Matriz Viral/metabolismo , Linhagem Celular , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza B/genética , Influenza Humana/enzimologia , Influenza Humana/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteínas da Matriz Viral/genética , Replicação Viral
4.
Molecules ; 26(23)2021 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-34885710

RESUMO

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.


Assuntos
Adenosina Trifosfatases/química , Endonucleases/química , Inibidores Enzimáticos/química , Influenza Humana/enzimologia , Adenosina Trifosfatases/antagonistas & inibidores , Adenosina Trifosfatases/ultraestrutura , Domínio Catalítico/efeitos dos fármacos , Desenho de Fármacos/tendências , Endonucleases/antagonistas & inibidores , Endonucleases/ultraestrutura , Humanos , Influenza Humana/tratamento farmacológico , Influenza Humana/virologia , Ligantes , Modelos Moleculares , Simulação de Acoplamento Molecular , Relação Quantitativa Estrutura-Atividade
5.
Future Microbiol ; 16: 445-452, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33847136

RESUMO

Influenza A virus (IAV) is a crucial cause of respiratory infections in humans worldwide. Therefore, studies should clarify adaptation mechanisms of IAV and critical factors of the viral pathogenesis in human hosts. GTPases of the Rab family are the largest branch of the Ras-like small GTPase superfamily, and they regulate almost every step during vesicle-mediated trafficking. Evidence has shown that Rab proteins participate in the lifecycle of IAV. In this mini-review, we outline the regulatory mechanisms of different Rab proteins in the lifecycle of IAV. Understanding the role of Rab proteins in IAV infections is important to develop broad-spectrum host-targeted antiviral strategies.


Assuntos
Vírus da Influenza A/fisiologia , Influenza Humana/virologia , Proteínas rab de Ligação ao GTP/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Influenza Humana/enzimologia , Ribonucleoproteínas/metabolismo , Proteínas Virais/metabolismo , Montagem de Vírus , Internalização do Vírus , Replicação Viral
6.
J Biol Chem ; 296: 100486, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33647314

RESUMO

Baloxavir marboxil (BXM) is an FDA-approved antiviral prodrug for the treatment of influenza A and B infection and postexposure prophylaxis. The active form, baloxavir acid (BXA), targets the cap-snatching endonuclease (PA) of the influenza virus polymerase complex. The nuclease activity delivers the primer for transcription, and previous reports have shown that BXA blocks the nuclease activity with high potency. However, biochemical studies on the mechanism of action are lacking. Structural data have shown that BXA chelates the two divalent metal ions at the active site, like inhibitors of the human immunodeficiency virus type 1 (HIV-1) integrase or ribonuclease (RNase) H. Here we studied the mechanisms underlying the high potency of BXA and how the I38T mutation confers resistance to the drug. Enzyme kinetics with the recombinant heterotrimeric enzyme (FluB-ht) revealed characteristics of a tight binding inhibitor. The apparent inhibitor constant (Kiapp) is 12 nM, while the I38T mutation increased Kiapp by ∼18-fold. Order-of-addition experiments show that a preformed complex of FluB-ht, Mg2+ ions and BXA is required to observe inhibition, which is consistent with active site binding. Conversely, a preformed complex of FluB-ht and RNA substrate prevents BXA from accessing the active site. Unlike integrase inhibitors that interact with the DNA substrate, BXA behaves like RNase H inhibitors that compete with the nucleic acid at the active site. The collective data support the conclusion that BXA is a tight binding inhibitor and the I38T mutation diminishes these properties.


Assuntos
Dibenzotiepinas/farmacologia , Endonucleases/antagonistas & inibidores , Vírus da Influenza B/efeitos dos fármacos , Influenza Humana/tratamento farmacológico , Influenza Humana/virologia , Morfolinas/farmacologia , Piridonas/farmacologia , RNA Polimerase Dependente de RNA/antagonistas & inibidores , Triazinas/farmacologia , Replicação Viral/efeitos dos fármacos , Antivirais/farmacologia , Domínio Catalítico , Endonucleases/metabolismo , Humanos , Vírus da Influenza B/enzimologia , Vírus da Influenza B/isolamento & purificação , Influenza Humana/enzimologia , RNA Polimerase Dependente de RNA/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo
7.
J Biol Chem ; 296: 100470, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33639165

RESUMO

The ongoing COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a major threat to global health. Vaccines are ideal solutions to prevent infection, but treatments are also needed for those who have contracted the virus to limit negative outcomes, when vaccines are not applicable. Viruses must cross host cell membranes during their life cycle, creating a dependency on processes involving membrane dynamics. Thus, in this study, we examined whether the synthetic machinery for glycosphingolipids, biologically active components of cell membranes, can serve as a therapeutic target to combat SARS-CoV-2. We examined the antiviral effect of two specific inhibitors of glucosylceramide synthase (GCS): (i) Genz-123346, an analogue of the United States Food and Drug Administration-approved drug Cerdelga and (ii) GENZ-667161, an analogue of venglustat, which is currently under phase III clinical trials. We found that both GCS inhibitors inhibit replication of SARS-CoV-2. Moreover, these inhibitors also disrupt replication of influenza virus A/PR/8/34 (H1N1). Our data imply that synthesis of glycosphingolipids is necessary to support viral life cycles and suggest that GCS inhibitors should be further explored as antiviral therapies.


Assuntos
Antivirais/farmacologia , Carbamatos/farmacologia , Dioxanos/farmacologia , Glucosiltransferases/antagonistas & inibidores , Glicoesfingolipídeos/antagonistas & inibidores , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Pirrolidinas/farmacologia , Quinuclidinas/farmacologia , SARS-CoV-2/efeitos dos fármacos , Animais , Antivirais/síntese química , COVID-19/enzimologia , COVID-19/virologia , Carbamatos/síntese química , Membrana Celular/efeitos dos fármacos , Membrana Celular/enzimologia , Membrana Celular/virologia , Chlorocebus aethiops , Ensaios Clínicos Fase III como Assunto , Dioxanos/síntese química , Cães , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica , Glucosiltransferases/genética , Glucosiltransferases/metabolismo , Glicoesfingolipídeos/biossíntese , Interações Hospedeiro-Patógeno/genética , Humanos , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H1N1/metabolismo , Influenza Humana/tratamento farmacológico , Influenza Humana/enzimologia , Influenza Humana/virologia , Células Madin Darby de Rim Canino , Pirrolidinas/síntese química , Quinuclidinas/síntese química , SARS-CoV-2/crescimento & desenvolvimento , SARS-CoV-2/metabolismo , Transdução de Sinais , Células Vero , Replicação Viral/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
8.
Oxid Med Cell Longev ; 2020: 8824934, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33204399

RESUMO

Antigenic mismatch can cause influenza vaccines to be ineffective, and influenza viruses resistant to antiviral drugs are rising. Thus, development of antiviral agents against these viruses is an immediate need. Rhus verniciflua (RVS) has long been used in herbal medicine and as a nutritional supplement. The effect of RVS and its components on influenza virus has not, however, been reported. We found that RVS treatment significantly reduced viral replication when evaluated with green fluorescent protein- (GFP-) tagged virus (influenza A virus, A/PR/8/34-GFP) in Madin-Darby canine kidney (MDCK) cells. RVS showed significant inhibition of neuraminidase from A/PR/8/34. Subsequently, three fractions were prepared from an ethanolic crude extract of RVS. In vitro assays indicated that an ethyl acetate fraction (RVSE) was more potent than H2O and CHCl3 fractions. RVSE significantly suppressed influenza virus infection in MDCK cells via neuraminidase inhibition. Additionally, RVSE treatment inhibited expression of several virus proteins and decreased mortality of mice exposed to influenza A/PR/8/34 by 50% and reduced weight loss by 11.5%. Active components in RVSE were isolated, and 5-deoxyluteolin (5) and sulfuretin (7) demonstrate the highest neuraminidase inhibitory activity against influenza A virus. RVS, RVSE, and their constituents may be useful for the development of anti-influenza agents.


Assuntos
Antivirais/farmacologia , Inibidores Enzimáticos/farmacologia , Influenza Humana/tratamento farmacológico , Neuraminidase/antagonistas & inibidores , Infecções por Orthomyxoviridae/tratamento farmacológico , Extratos Vegetais/farmacologia , Rhus/química , Células A549 , Acetatos/química , Animais , Cães , Etanol/química , Feminino , Humanos , Vírus da Influenza A/efeitos dos fármacos , Influenza Humana/enzimologia , Influenza Humana/virologia , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/enzimologia , Infecções por Orthomyxoviridae/virologia , Fitoterapia , Proteínas Virais/metabolismo , Replicação Viral
9.
J Biol Chem ; 295(33): 11388-11407, 2020 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-32303635

RESUMO

Cleavage of influenza virus hemagglutinin (HA) by host proteases is essential for virus infectivity. HA of most influenza A and B (IAV/IBV) viruses is cleaved at a monobasic motif by trypsin-like proteases. Previous studies have reported that transmembrane serine protease 2 (TMPRSS2) is essential for activation of H7N9 and H1N1pdm IAV in mice but that H3N2 IAV and IBV activation is independent of TMPRSS2 and carried out by as-yet-undetermined protease(s). Here, to identify additional H3 IAV- and IBV-activating proteases, we used RNA-Seq to investigate the protease repertoire of murine lower airway tissues, primary type II alveolar epithelial cells (AECIIs), and the mouse lung cell line MLE-15. Among 13 candidates identified, TMPRSS4, TMPRSS13, hepsin, and prostasin activated H3 and IBV HA in vitro IBV activation and replication was reduced in AECIIs from Tmprss2/Tmprss4-deficient mice compared with WT or Tmprss2-deficient mice, indicating that murine TMPRSS4 is involved in IBV activation. Multicycle replication of H3N2 IAV and IBV in AECIIs of Tmprss2/Tmprss4-deficient mice varied in sensitivity to protease inhibitors, indicating that different, but overlapping, sets of murine proteases facilitate H3 and IBV HA cleavages. Interestingly, human hepsin and prostasin orthologs did not activate H3, but they did activate IBV HA in vitro Our results indicate that TMPRSS4 is an IBV-activating protease in murine AECIIs and suggest that TMPRSS13, hepsin, and prostasin cleave H3 and IBV HA in mice. They further show that hepsin and prostasin orthologs might contribute to the differences observed in TMPRSS2-independent activation of H3 in murine and human airways.


Assuntos
Vírus da Influenza A Subtipo H3N2/fisiologia , Vírus da Influenza B/fisiologia , Influenza Humana/enzimologia , Infecções por Orthomyxoviridae/enzimologia , Peptídeo Hidrolases/metabolismo , Ativação Viral , Animais , Linhagem Celular , Cães , Ativação Enzimática/efeitos dos fármacos , Perfilação da Expressão Gênica , Células HEK293 , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Vírus da Influenza A Subtipo H3N2/efeitos dos fármacos , Vírus da Influenza B/efeitos dos fármacos , Influenza Humana/tratamento farmacológico , Influenza Humana/genética , Influenza Humana/virologia , Pulmão/enzimologia , Pulmão/metabolismo , Pulmão/virologia , Células Madin Darby de Rim Canino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/virologia , Peptídeo Hidrolases/genética , Inibidores de Proteases/farmacologia , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Ativação Viral/efeitos dos fármacos
10.
J Med Chem ; 63(8): 4205-4214, 2020 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-32227946

RESUMO

Influenza and dengue viruses present a growing global threat to public health. Both viruses depend on the host endoplasmic reticulum (ER) glycoprotein folding pathway. In 2014, Sadat et al. reported two siblings with a rare genetic defect in ER α-glucosidase I (ER Glu I) who showed resistance to viral infections, identifying ER Glu I as a key antiviral target. Here, we show that a single dose of UV-4B (the hydrochloride salt form of N-(9'-methoxynonyl)-1-deoxynojirimycin; MON-DNJ) capable of inhibiting Glu I in vivo is sufficient to prevent death in mice infected with lethal viral doses, even when treatment is started as late as 48 h post infection. The first crystal structure of mammalian ER Glu I will constitute the basis for the development of potent and selective inhibitors. Targeting ER Glu I with UV-4B-derived compounds may alter treatment paradigms for acute viral disease through development of a single-dose therapeutic regime.


Assuntos
Dengue/prevenção & controle , Retículo Endoplasmático/efeitos dos fármacos , Inibidores de Glicosídeo Hidrolases/administração & dosagem , Influenza Humana/prevenção & controle , alfa-Glucosidases , Animais , Dengue/tratamento farmacológico , Dengue/enzimologia , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/enzimologia , Relação Dose-Resposta a Droga , Retículo Endoplasmático/enzimologia , Humanos , Influenza Humana/tratamento farmacológico , Influenza Humana/enzimologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Estrutura Secundária de Proteína , alfa-Glucosidases/metabolismo
11.
Eur J Health Econ ; 21(5): 775-785, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32180069

RESUMO

OBJECTIVE: Influenza virus infections cause a high disease and economic burden during seasonal epidemics. However, there is still a need for reliable disease burden estimates to provide a more detailed picture of the impact of influenza. Therefore, the objectives of this study is to estimate the incidence of hospitalisation for influenza virus infection and associated hospitalisation costs in adult patients in the Netherlands during two consecutive influenza seasons. METHODS: We conducted a retrospective study in adult patients with a laboratory confirmed influenza virus infection in three Dutch hospitals during respiratory seasons 2014-2015 and 2015-2016. Incidence was calculated as the weekly number of hospitalised influenza patients divided by the total population in the catchment populations of the three hospitals. Arithmetic mean hospitalisation costs per patient were estimated and included costs for emergency department consultation, diagnostics, general ward and/or intensive care unit admission, isolation, antibiotic and/or antiviral treatment. These hospitalisation costs were extrapolated to national level and expressed in 2017 euros. RESULTS: The study population consisted of 380 hospitalised adult influenza patients. The seasonal cumulative incidence was 3.5 cases per 10,000 persons in respiratory season 2014-2015, compared to 1.8 cases per 10,000 persons in 2015-2016. The arithmetic mean hospitalisation cost per influenza patient was €6128 (95% CI €4934-€7737) per patient in 2014-2015 and €8280 (95% CI €6254-€10,665) in 2015-2016, potentially reaching total hospitalisation costs of €28 million in 2014-2015 and €20 million in 2015-2016. CONCLUSIONS: Influenza virus infections lead to 1.8-3.5 hospitalised patients per 10,000 persons, with mean hospitalisation costs of €6100-€8300 per adult patient, resulting in 20-28 million euros annually in The Netherlands. The highest arithmetic mean hospitalisation costs per patient were found in the 45-64 year age group. These influenza burden estimates could be used for future influenza cost-effectiveness and impact studies.


Assuntos
Custos Hospitalares/estatística & dados numéricos , Hospitalização/economia , Influenza Humana/economia , Influenza Humana/epidemiologia , Adolescente , Adulto , Idoso , Feminino , Hospitalização/estatística & dados numéricos , Humanos , Incidência , Vírus da Influenza A/isolamento & purificação , Influenza Humana/enzimologia , Masculino , Pessoa de Meia-Idade , Países Baixos/epidemiologia , Estudos Retrospectivos , Adulto Jovem
12.
Cell Mol Biol Lett ; 25: 15, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32161622

RESUMO

Based on available metabolomic studies, influenza infection affects a variety of cellular metabolic pathways to ensure an optimal environment for its replication and production of viral particles. Following infection, glucose uptake and aerobic glycolysis increase in infected cells continually, which results in higher glucose consumption. The pentose phosphate shunt, as another glucose-consuming pathway, is enhanced by influenza infection to help produce more nucleotides, especially ATP. Regarding lipid species, following infection, levels of triglycerides, phospholipids, and several lipid derivatives undergo perturbations, some of which are associated with inflammatory responses. Also, mitochondrial fatty acid ß-oxidation decreases significantly simultaneously with an increase in biosynthesis of fatty acids and membrane lipids. Moreover, essential amino acids are demonstrated to decline in infected tissues due to the production of large amounts of viral and cellular proteins. Immune responses against influenza infection, on the other hand, could significantly affect metabolic pathways. Mainly, interferon (IFN) production following viral infection affects cell function via alteration in amino acid synthesis, membrane composition, and lipid metabolism. Understanding metabolic alterations required for influenza virus replication has revealed novel therapeutic methods based on targeted inhibition of these cellular metabolic pathways.


Assuntos
Metabolismo Energético , Glucose/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Influenza Humana/metabolismo , Interferons/metabolismo , Metabolismo dos Lipídeos , Metabolismo Energético/genética , Ácido Graxo Sintases/metabolismo , Glicólise , Interações Hospedeiro-Patógeno/genética , Humanos , Influenza Humana/enzimologia , Influenza Humana/imunologia , Influenza Humana/terapia , Interferons/imunologia , Metabolismo dos Lipídeos/genética , Redes e Vias Metabólicas/genética , Óxido Nítrico/metabolismo
13.
Curr Top Med Chem ; 20(23): 2046-2055, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31738149

RESUMO

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.


Assuntos
Antivirais/farmacologia , Cinamatos/farmacologia , Depsídeos/farmacologia , Inibidores Enzimáticos/farmacologia , Influenza Humana/tratamento farmacológico , Simulação de Dinâmica Molecular , Neuraminidase/antagonistas & inibidores , Antivirais/química , Ácidos Cafeicos/química , Ácidos Cafeicos/farmacologia , Cinamatos/química , Depsídeos/química , Inibidores Enzimáticos/química , Humanos , Influenza Humana/enzimologia , Ligantes , Estrutura Molecular , Neuraminidase/metabolismo , Termodinâmica , Ácido Rosmarínico
14.
Influenza Other Respir Viruses ; 13(6): 603-609, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31489989

RESUMO

BACKGROUND: Seasonal influenza is an important cause of morbidity and mortality worldwide. Immune activation after stimulation with interferon-gamma leads to increased production of neopterin but also results in increased tryptophan catabolism through indoleamine 2,3-dioxygenase (IDO). Our pilot study determined neopterin serum levels and IDO activity in patients with influenza infection and investigated whether neopterin is linked to clinical outcome parameters (mortality ≤30 days, acute cardiac events (ACE) length of hospitalization, ICU admission). METHODS: Neopterin concentrations were analyzed in serum samples of 40 patients with a confirmed diagnosis of influenza infection and in-hospital treatment for >24 hours. Data were compared to values of 100 healthy blood donors and 48 age-matched pneumonia patients. In a subgroup of 14 patients, tryptophan and kynurenine concentrations, as well as kynurenine-to-tryptophan ratio, were analyzed. RESULTS: In all influenza patients, neopterin concentrations were increased and significantly higher compared to those determined in patients with pneumonia and healthy controls. Positive correlations between the duration of hospitalization and neopterin were found. Significantly higher levels of kynurenine, kynurenine-to-tryptophan ratio, and lower levels of tryptophan were seen in influenza patients compared to healthy controls. CONCLUSIONS: Neopterin seems to be related to the course of the disease and could be a valuable biomarker to identify patients at an elevated risk of a worsened outcome; however, further prospective validation studies are needed to support the here presented preliminary results.


Assuntos
Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Influenza Humana/sangue , Neopterina/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/sangue , Feminino , Hospitalização , Humanos , Influenza Humana/enzimologia , Cinurenina/sangue , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Pneumonia/sangue , Pneumonia/enzimologia , Curva ROC , Estudos Retrospectivos , Triptofano/sangue
15.
J Virol ; 93(21)2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31391268

RESUMO

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.


Assuntos
Vírus da Influenza A/fisiologia , Vírus da Influenza B/fisiologia , Influenza Humana/virologia , Serina Endopeptidases/metabolismo , Animais , Brônquios/citologia , Células Cultivadas , Células Epiteliais/virologia , Técnicas de Silenciamento de Genes , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Influenza Humana/enzimologia , Influenza Humana/metabolismo , Camundongos , Infecções por Orthomyxoviridae/enzimologia , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Alvéolos Pulmonares/citologia , Serina Endopeptidases/genética , Regulação para Cima , Replicação Viral
16.
Virology ; 534: 80-86, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31220651

RESUMO

The immunopathological mechanisms as well as the role played by influenza A virus infection of human leukocytes and induction of apoptosis have not been fully elucidated. We confirm here that the percentage of cells that are infected is less than the percent of apoptotic cells. Depletion of monocytes/macrophages and depletion of cells expressing influenza neuraminidase from the cultures after exposure to virus decreased lymphocyte apoptosis. Treatment of virus-exposed leukocyte cultures with anti-neuraminidase antibodies but not with anti-hemagglutinin antibodies, reduced lymphocyte production of active caspase-3 and induction of apoptosis. Different strains of virus induced different levels of apoptosis. Variations in induction of apoptosis correlated with production and expression of viral neuraminidase by infected leukocytes. The data suggest that cell surface expression of neuraminidase plays an important role in the induction of apoptosis in human lymphocytes. The benefit, or cost, to the host of lymphocyte apoptosis warrants continued investigation.


Assuntos
Apoptose , Membrana Celular/virologia , Vírus da Influenza A/enzimologia , Influenza Humana/virologia , Linfócitos/citologia , Neuraminidase/metabolismo , Proteínas Virais/metabolismo , Animais , Caspase 3/genética , Caspase 3/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A/genética , Influenza Humana/enzimologia , Influenza Humana/genética , Influenza Humana/fisiopatologia , Linfócitos/virologia , Neuraminidase/genética , Proteínas Virais/genética
17.
Virology ; 534: 54-63, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31176924

RESUMO

Influenza A virus (IAV) infections result in ∼500,000 global deaths annually. Host kinases link multiple signaling pathways at various stages of infection and are attractive therapeutic target. Focal adhesion kinase (FAK), a non-receptor tyrosine kinase, regulates several cellular processes including NFkB and antiviral responses. We investigated how FAK kinase activity regulates IAV pathogenesis. Using a severe infection model, we infected IAV-susceptible DBA/2 J mice with a lethal dose of H1N1 IAV. We observed reduced viral load and pro-inflammatory cytokines, delayed mortality, and increased survival in FAK inhibitor (Y15) treated mice. In vitro IAV-induced NFkB-promoter activity was reduced by Y15 or a dominant negative kinase-dead FAK mutant (FAK-KD) independently of the viral immune modulator, NS1. Finally, we observed reduced IAV-induced nuclear localization of NFkB in FAK-KD expressing cells. Our data suggest a novel mechanism where IAV hijacks FAK to promote viral replication and limit its ability to contribute to innate immune responses.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/imunologia , Vírus da Influenza A Subtipo H1N1/patogenicidade , Influenza Humana/enzimologia , NF-kappa B/imunologia , Provírus/patogenicidade , Animais , Feminino , Proteína-Tirosina Quinases de Adesão Focal/genética , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/fisiologia , Influenza Humana/genética , Influenza Humana/imunologia , Influenza Humana/virologia , Camundongos , Camundongos Endogâmicos DBA , NF-kappa B/genética , Regiões Promotoras Genéticas , Provírus/genética , Provírus/fisiologia , Carga Viral , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Virulência , Replicação Viral
18.
J Gen Virol ; 100(7): 1073-1078, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31099738

RESUMO

The surface protein haemagglutinin (HA) of influenza A viruses (IAV) needs to be cleaved by a host protease to become functional. Here, we investigated if IAV of the H10 subtype also requires TMPRSS2 for replication and pathogenesis in mice. We first showed in cell culture that TMPRSS2 is able to cleave H10-HA. When Tmprss2-/- deficient mice were infected with a re-assorted virus H10-HA, they did not lose body weight and no viral replication was observed in contrast to wild-type mice. Histopathological analysis showed that inflammatory lesions in the lung of Tmprss2-/- mice were reduced compared to wild-type mice. In addition, no viral antigen was detected in the lungs of Tmprss2-/- mice and no evidence for HA cleavage was observed. We conclude from these studies that TMPRSS2 activity is also essential for in vivo replication and pathogenesis of H10 IAV.


Assuntos
Vírus da Influenza A/fisiologia , Influenza Humana/enzimologia , Serina Endopeptidases/genética , Animais , Feminino , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A/genética , Vírus da Influenza A/patogenicidade , Influenza Humana/genética , Influenza Humana/imunologia , Influenza Humana/virologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Serina Endopeptidases/deficiência , Virulência , Replicação Viral
19.
Viruses ; 11(2)2019 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-30791550

RESUMO

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.


Assuntos
Interações Hospedeiro-Patógeno , Influenza Humana/enzimologia , Fosfotransferases/metabolismo , Internalização do Vírus , Antivirais/uso terapêutico , Linhagem Celular , Humanos , Quinase I-kappa B/metabolismo , Imunidade Inata , Vírus da Influenza A Subtipo H1N1 , Virus da Influenza A Subtipo H5N1 , Subtipo H7N9 do Vírus da Influenza A , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
J Infect Dis ; 219(2): 177-185, 2019 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-30053042

RESUMO

Background: AL-794 is an orally active prodrug of ALS-033719, which selectively inhibits the endonuclease domain of influenza virus A and B polymerase. Methods: In a phase 1, double-blinded, randomized, placebo-controlled study, healthy subjects were inoculated intranasally with influenza virus (A/Perth/16/2009 H3N2) after confirmation of infection or on day 4. Subjects received 50 mg of AL-794, 150 mg of AL-794, or placebo twice daily for 5 days. Viral load, influenza symptoms, pharmacokinetics, and safety were evaluated. Results: A total of 61 subjects were inoculated. In 42 infected subjects, the mean peak viral load for 50-mg AL-794 recipients, 150-mg AL-794 recipients, and placebo recipients was 3.54, 2.77, and 3.72 log10 50% tissue culture infectious doses (TCID50)/mL, respectively. The mean influenza viral load area under the curve in the corresponding treatment groups was 137, 87.5, and 142 log10 TCID50/mL·h, respectively, and the median time to virus nondetection was 117, 75.3, and 108 hours, respectively. AL-794 was well tolerated, and no viral resistance to ALS-033719 was identified. Conclusion: Following oral administration of AL-794, significant dose-dependent antiviral activity was noted, with a greater decrease in viral load, symptoms, and mucus weight at the 150-mg dose, compared with the 50-mg dose, and no safety concerns for either dose or placebo. Clinical Trials Registration: NCT02588521.


Assuntos
Antivirais/farmacologia , Antivirais/uso terapêutico , Endonucleases/antagonistas & inibidores , Influenza Humana/tratamento farmacológico , Serina Endopeptidases/farmacologia , Serina Endopeptidases/uso terapêutico , Administração Oral , Adolescente , Adulto , Antivirais/efeitos adversos , Método Duplo-Cego , Feminino , Humanos , Vírus da Influenza A Subtipo H3N2 , Influenza Humana/enzimologia , Masculino , Pessoa de Meia-Idade , Carga Viral/efeitos dos fármacos , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...