Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
EMBO Rep ; 23(11): e51709, 2022 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-36094794

RESUMEN

Phosphatidylserine (PS) is a critical lipid factor in the assembly and spread of numerous lipid-enveloped viruses. Here, we describe the ability of the Ebola virus (EBOV) matrix protein eVP40 to induce clustering of PS and promote viral budding in vitro, as well as the ability of an FDA-approved drug, fendiline, to reduce PS clustering and subsequent virus budding and entry. To gain mechanistic insight into fendiline inhibition of EBOV replication, multiple in vitro assays were run including imaging, viral budding and viral entry assays. Fendiline lowers PS content in mammalian cells and PS in the plasma membrane, where the ability of VP40 to form new virus particles is greatly lower. Further, particles that form from fendiline-treated cells have altered particle morphology and cannot significantly infect/enter cells. These complementary studies reveal the mechanism by which EBOV matrix protein clusters PS to enhance viral assembly, budding, and spread from the host cell while also laying the groundwork for fundamental drug targeting strategies.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , Animales , Fiebre Hemorrágica Ebola/metabolismo , Ebolavirus/fisiología , Fosfatidilserinas/metabolismo , Fendilina/metabolismo , Proteínas de la Matriz Viral/metabolismo , Ensamble de Virus , Análisis por Conglomerados , Mamíferos/metabolismo
2.
Biol Pharm Bull ; 44(12): 1837-1842, 2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-34615812

RESUMEN

Epstein-Barr virus (EBV), a human herpesvirus, is several human lymphoid malignancies-associated. Our earlier study found the effect of Polygonum cuspidatum root on promoting EBV-positive apoptosis. Therefore, this study investigated the effects of the Polygonum cuspidatum ethyl acetate subfraction containing emodin on EBV gene expression and anti-EBV tumor cells. Resultantly, the the Polygonum cuspidatum ethyl acetate subfraction containing emodin (F3a) promoted Raji cell death (50% cytotoxic concentration, CC50: 12.08 µg/mL); the 12.5 µg/mL F3a effect transcribed BRLF1 and BNLF1 and increased latent membrane protein 1 (LMP1), which may reduce the intracellular phospho-extracellular signal-regulated kinase (ERK) and phospho-inhibitor of Nuclear factor kappa B α (IκBα). Meanwhile, the Raji cells increased the intracellular reactive-oxygen species (ROS), activated the apoptosis-related proteins, cleaved caspase 3 and poly(ADP-ribose)polymerase (PARP), and increased the apoptosis percentage. Therefore, the Polygonum cuspidatum ethyl acetate subfraction containing emodin could be a therapeutic drug for EBV-related tumors.


Asunto(s)
Emodina/farmacología , Infecciones por Virus de Epstein-Barr/metabolismo , Fallopia japonica/química , Herpesvirus Humano 4/metabolismo , Neoplasias/virología , Extractos Vegetales/farmacología , Proteínas Virales/metabolismo , Antineoplásicos Fitogénicos/farmacología , Antineoplásicos Fitogénicos/uso terapéutico , Apoptosis , Linfoma de Burkitt/virología , Línea Celular Tumoral , Emodina/uso terapéutico , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/virología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Expresión Génica , Humanos , Inhibidor NF-kappaB alfa/metabolismo , FN-kappa B/metabolismo , Fitoterapia , Extractos Vegetales/uso terapéutico , Raíces de Plantas/química , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Proteínas de la Matriz Viral/metabolismo
3.
Sci Rep ; 11(1): 19223, 2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34584169

RESUMEN

Respiratory syncytial virus (RSV) is the primary cause of serious lower respiratory tract disease in infants, young children, the elderly and immunocompromised individuals. Therapy for RSV infections is limited to high risk infants and there are no safe and efficacious vaccines. Matrix (M) protein is a major RSV structural protein with a key role in virus assembly. Interestingly, M is localised to the nucleus early in infection and its export into the cytoplasm by the nuclear exporter, exportin-1 (XPO1) is essential for RSV assembly. We have shown previously that chemical inhibition of XPO1 function results in reduced RSV replication. In this study, we have investigated the anti-RSV efficacy of Selective Inhibitor of Nuclear Export (SINE) compounds, KPT-335 and KPT-185. Our data shows that therapeutic administration of the SINE compounds results in reduced RSV titre in human respiratory epithelial cell culture. Within 24 h of treatment, RSV replication and XPO1 expression was reduced, M protein was partially retained in the nucleus, and cell cycle progression was delayed. Notably, the effect of SINE compounds was reversible within 24 h after their removal. Our data show that reversible inhibition of XPO1 can disrupt RSV replication by affecting downstream pathways regulated by the nuclear exporter.


Asunto(s)
Acrilatos/farmacología , Carioferinas/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Triazoles/farmacología , Proteínas de la Matriz Viral/metabolismo , Replicación Viral/efectos de los fármacos , Células A549 , Acrilatos/uso terapéutico , Núcleo Celular/metabolismo , Evaluación Preclínica de Medicamentos , Humanos , Carioferinas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Virus Sincitial Respiratorio Humano/metabolismo , Triazoles/uso terapéutico , Proteína Exportina 1
4.
Eur J Med Chem ; 225: 113789, 2021 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-34438124

RESUMEN

SARS-CoV-2 as a positive-sense single-stranded RNA coronavirus caused the global outbreak of COVID-19. The main protease (Mpro) of the virus as the major enzyme processing viral polyproteins contributed to the replication and transcription of SARS-CoV-2 in host cells, and has been characterized as an attractive target in drug discovery. Herein, a set of 1,4-naphthoquinones with juglone skeleton were prepared and evaluated for the inhibitory efficacy against SARS-CoV-2 Mpro. More than half of the tested naphthoquinones could effectively inhibit the target enzyme with an inhibition rate of more than 90% at the concentration of 10 µM. In the structure-activity relationships (SARs) analysis, the characteristics of substituents and their position on juglone core scaffold were recognized as key ingredients for enzyme inhibitory activity. The most active compound, 2-acetyl-8-methoxy-1,4-naphthoquinone (15), which exhibited much higher potency in enzyme inhibitions than shikonin as the positive control, displayed an IC50 value of 72.07 ± 4.84 nM towards Mpro-mediated hydrolysis of the fluorescently labeled peptide. It fit well into the active site cavity of the enzyme by forming hydrogen bonds with adjacent amino acid residues in molecular docking studies. The results from in vitro antiviral activity evaluation demonstrated that the most potent Mpro inhibitor could significantly suppress the replication of SARS-CoV-2 in Vero E6 cells within the low micromolar concentrations, with its EC50 value of about 4.55 µM. It was non-toxic towards the host Vero E6 cells under tested concentrations. The present research work implied that juglone skeleton could be a primary template for the development of potent Mpro inhibitors.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Naftoquinonas/química , Inhibidores de Proteasas/uso terapéutico , SARS-CoV-2/enzimología , Proteínas de la Matriz Viral/antagonistas & inhibidores , Animales , Sitios de Unión , COVID-19/patología , COVID-19/virología , Dominio Catalítico , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Humanos , Enlace de Hidrógeno , Simulación del Acoplamiento Molecular , Naftoquinonas/metabolismo , Naftoquinonas/farmacología , Naftoquinonas/uso terapéutico , Inhibidores de Proteasas/química , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasas/farmacología , SARS-CoV-2/aislamiento & purificación , Relación Estructura-Actividad , Células Vero , Proteínas de la Matriz Viral/metabolismo
5.
Int J Mol Sci ; 22(13)2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-34208928

RESUMEN

The development of new antiviral drugs against SARS-CoV-2 is a valuable long-term strategy to protect the global population from the COVID-19 pandemic complementary to the vaccination. Considering this, the viral main protease (Mpro) is among the most promising molecular targets in light of its importance during the viral replication cycle. The natural flavonoid quercetin 1 has been recently reported to be a potent Mpro inhibitor in vitro, and we explored the effect produced by the introduction of organoselenium functionalities in this scaffold. In particular, we report here a new synthetic method to prepare previously inaccessible C-8 seleno-quercetin derivatives. By screening a small library of flavonols and flavone derivatives, we observed that some compounds inhibit the protease activity in vitro. For the first time, we demonstrate that quercetin (1) and 8-(p-tolylselenyl)quercetin (2d) block SARS-CoV-2 replication in infected cells at non-toxic concentrations, with an IC50 of 192 µM and 8 µM, respectively. Based on docking experiments driven by experimental evidence, we propose a non-covalent mechanism for Mpro inhibition in which a hydrogen bond between the selenium atom and Gln189 residue in the catalytic pocket could explain the higher Mpro activity of 2d and, as a result, its better antiviral profile.


Asunto(s)
Antivirales/química , Quercetina/química , SARS-CoV-2/metabolismo , Selenio/química , Proteínas de la Matriz Viral/antagonistas & inhibidores , Animales , Antivirales/metabolismo , Antivirales/farmacología , Sitios de Unión , COVID-19/patología , COVID-19/virología , Dominio Catalítico , Chlorocebus aethiops , Humanos , Enlace de Hidrógeno , Simulación del Acoplamiento Molecular , Inhibidores de Proteasas/química , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasas/farmacología , Quercetina/metabolismo , Quercetina/farmacología , SARS-CoV-2/aislamiento & purificación , Selenio/metabolismo , Células Vero , Proteínas de la Matriz Viral/metabolismo , Replicación Viral/efectos de los fármacos
6.
Int J Biol Macromol ; 184: 297-312, 2021 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-34118289

RESUMEN

COVID-19 caused by SARS-CoV-2 corona virus has become a global pandemic. In the absence of drugs and vaccine, and premises of time, efforts and cost required for their development, natural resources such as herbs are anticipated to provide some help and may also offer a promising resource for drug development. Here, we have investigated the therapeutic prospective of Ashwagandha for the COVID-19 pandemic. Nine withanolides were tested in silico for their potential to target and inhibit (i) cell surface receptor protein (TMPRSS2) that is required for entry of virus to host cells and (ii) viral protein (the main protease Mpro) that is essential for virus replication. We report that the withanolides possess capacity to inhibit the activity of TMPRSS2 and Mpro. Furthermore, withanolide-treated cells showed downregulation of TMPRSS2 expression and inhibition of SARS-CoV-2 replication in vitro, suggesting that Ashwagandha may provide a useful resource for COVID-19 treatment.


Asunto(s)
Antivirales/farmacología , Extractos Vegetales/química , SARS-CoV-2/fisiología , Serina Endopeptidasas/metabolismo , Proteínas de la Matriz Viral/metabolismo , Witanólidos/farmacología , Células A549 , Antivirales/química , Línea Celular , Supervivencia Celular/efectos de los fármacos , Simulación por Computador , Regulación hacia Abajo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7 , Modelos Moleculares , Simulación de Dinámica Molecular , Conformación Proteica , SARS-CoV-2/efectos de los fármacos , Serina Endopeptidasas/química , Proteínas de la Matriz Viral/química , Internalización del Virus/efectos de los fármacos , Witanólidos/química
7.
Molecules ; 26(8)2021 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-33921289

RESUMEN

The recent coronavirus disease 2019 (COVID-19) pandemic is a global threat for healthcare management and the economic system, and effective treatments against the pathogenic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus responsible for this disease have not yet progressed beyond the developmental phases. As drug refinement and vaccine progression require enormously broad investments of time, alternative strategies are urgently needed. In this study, we examined phytochemicals extracted from Avicennia officinalis and evaluated their potential effects against the main protease of SARS-CoV-2. The antioxidant activities of A. officinalis leaf and fruit extracts at 150 µg/mL were 95.97% and 92.48%, respectively. Furthermore, both extracts displayed low cytotoxicity levels against Artemia salina. The gas chromatography-mass spectroscopy analysis confirmed the identifies of 75 phytochemicals from both extracts, and four potent compounds, triacontane, hexacosane, methyl linoleate, and methyl palminoleate, had binding free energy values of -6.75, -6.7, -6.3, and -6.3 Kcal/mol, respectively, in complexes with the SARS-CoV-2 main protease. The active residues Cys145, Met165, Glu166, Gln189, and Arg188 in the main protease formed non-bonded interactions with the screened compounds. The root-mean-square difference (RMSD), root-mean-square fluctuations (RMSF), radius of gyration (Rg), solvent-accessible surface area (SASA), and hydrogen bond data from a molecular dynamics simulation study confirmed the docked complexes' binding rigidity in the atomistic simulated environment. However, this study's findings require in vitro and in vivo validation to ensure the possible inhibitory effects and pharmacological efficacy of the identified compounds.


Asunto(s)
Avicennia/química , Tratamiento Farmacológico de COVID-19 , Fitoquímicos/uso terapéutico , SARS-CoV-2/metabolismo , Antioxidantes/química , Antioxidantes/metabolismo , Antioxidantes/uso terapéutico , Avicennia/metabolismo , Sitios de Unión , COVID-19/patología , COVID-19/virología , Frutas/química , Frutas/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Alcohol Feniletílico/química , Alcohol Feniletílico/metabolismo , Alcohol Feniletílico/uso terapéutico , Fenilpropionatos/química , Fenilpropionatos/metabolismo , Fenilpropionatos/uso terapéutico , Fitoquímicos/química , Fitoquímicos/metabolismo , Hojas de la Planta/química , Hojas de la Planta/metabolismo , SARS-CoV-2/aislamiento & purificación , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/metabolismo
8.
BMC Complement Altern Med ; 19(1): 346, 2019 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-31791311

RESUMEN

BACKGROUND: Influenza A virus (IAV) is still a major health threat. The clinical manifestations of this infection are related to immune dysregulation, which causes morbidity and mortality. The usage of traditional medication with immunomodulatory properties against influenza infection has been increased recently. Our previous study showed antiviral activity of quercetin-3-O-α-L-rhamnopyranoside (Q3R) isolated from Rapanea melanophloeos (RM) (L.) Mez (family Myrsinaceae) against H1N1 (A/PR/8/34) infection. This study aimed to confirm the wider range of immunomodulatory effect of Q3R on selective pro- and anti-inflammatory cytokines against IAV in vitro, to evaluate the effect of Q3R on apoptosis pathway in combination with H1N1, also to assess the physical interaction of Q3R with virus glycoproteins and RhoA protein using computational docking. METHODS: MDCK cells were exposed to Q3R and 100CCID50/100 µl of H1N1 in combined treatments (co-, pre- and post-penetration treatments). The treatments were tested for the cytokines evaluation at RNA and protein levels by qPCR and ELISA, respectively. In another set of treatment, apoptosis was examined by detecting RhoA GTPase protein and caspase-3 activity. Molecular docking was used as a tool for evaluation of the potential anti-influenza activity of Q3R. RESULTS: The expressions of cytokines in both genome and protein levels were significantly affected by Q3R treatment. It was shown that Q3R was much more effective against influenza when it was applied in co-penetration treatment. Q3R in combination with H1N1 increased caspase-3 activity while decreasing RhoA activation. The molecular docking results showed strong binding ability of Q3R with M2 transmembrane, Neuraminidase of 2009 pandemic H1N1, N1 and H1 of PR/8/1934 and Human RhoA proteins, with docking energy of - 10.81, - 10.47, - 9.52, - 9.24 and - 8.78 Kcal/mol, respectively. CONCLUSIONS: Quercetin-3-O-α-L-rhamnopyranoside from RM was significantly effective against influenza infection by immunomodulatory properties, affecting the apoptosis pathway and binding ability to viral receptors M2 transmembrane and Neuraminidase of 2009 pandemic H1N1 and human RhoA cellular protein. Further research will focus on detecting the detailed specific mechanism of Q3R in virus-host interactions.


Asunto(s)
Antivirales , Glicósidos , Subtipo H1N1 del Virus de la Influenza A , Myrsine/química , Fitoquímicos , Quercetina/análogos & derivados , Animales , Antivirales/química , Antivirales/metabolismo , Antivirales/farmacología , Apoptosis/efectos de los fármacos , Citocinas/metabolismo , Perros , Glicósidos/química , Glicósidos/metabolismo , Glicósidos/farmacología , Células de Riñón Canino Madin Darby , Simulación del Acoplamiento Molecular , Neuraminidasa/química , Neuraminidasa/metabolismo , Fitoquímicos/química , Fitoquímicos/metabolismo , Fitoquímicos/farmacología , Quercetina/química , Quercetina/metabolismo , Quercetina/farmacología , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/metabolismo
9.
Am J Chin Med ; 47(6): 1307-1324, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31505936

RESUMEN

Aloe vera ethanol extract (AVE) reportedly has significant anti-influenza virus activity, but its underlying mechanisms of action and constituents have not yet been completely elucidated. Previously, we have confirmed that AVE treatment significantly reduces the viral replication of green fluorescent protein-labeled influenza A virus in Madin-Darby canine kidney (MDCK) cells. In addition, post-treatment with AVE inhibited viral matrix protein 1 (M1), matrix protein 2 (M2), and hemagglutinin (HA) mRNA synthesis and viral protein (M1, M2, and HA) expressions. In this study, we demonstrated that AVE inhibited autophagy induced by influenza A virus in MDCK cells and also identified quercetin, catechin hydrate, and kaempferol as the active antiviral components of AVE. We also found that post-treatment with quercetin, catechin hydrate, and kaempferol markedly inhibited M2 viral mRNA synthesis and M2 protein expression. A docking simulation suggested that the binding affinity of quercetin, catechin hydrate, and kaempferol for the M2 protein may be higher than that of known M2 protein inhibitors. Thus, the inhibition of autophagy induced by influenza virus may explain the antiviral activity of AVE against H1N1 or H3N2. Aloe vera extract and its constituents may, therefore, be potentially useful for the development of anti-influenza agents.


Asunto(s)
Aloe/química , Antivirales , Autofagia/efectos de los fármacos , Virus de la Influenza A/fisiología , Virus de la Influenza A/patogenicidad , Extractos Vegetales/farmacología , Replicación Viral/efectos de los fármacos , Animales , Células Cultivadas , Perros , Hemaglutininas Virales/genética , Hemaglutininas Virales/metabolismo , Subtipo H1N1 del Virus de la Influenza A , Subtipo H3N2 del Virus de la Influenza A , Virus de la Influenza A/metabolismo , Riñón/citología , Unión Proteica/efectos de los fármacos , Quercetina/metabolismo , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Proteínas de la Matriz Viral/metabolismo
10.
J Virol ; 92(24)2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30282713

RESUMEN

Screening of chemical libraries with 2,000 synthetic compounds identified salinomycin as a hit against influenza A and B viruses, with 50% effective concentrations ranging from 0.4 to 4.3 µM in cells. This compound is a carboxylic polyether ionophore that exchanges monovalent ions for protons across lipid bilayer membranes. Monitoring the time course of viral infection showed that salinomycin blocked nuclear migration of viral nuclear protein (NP), the most abundant component of the viral ribonucleoprotein (vRNP) complex. It caused cytoplasmic accumulation of NP, particularly within perinuclear endosomes, during virus entry. This was primarily associated with failure to acidify the endosomal-lysosomal compartments. Similar to the case with amantadine (AMT), proton channel activity of viral matrix protein 2 (M2) was blocked by salinomycin. Using purified retroviral Gag-based virus-like particles (VLPs) with M2, it was proved that salinomycin directly affects the kinetics of a proton influx into the particles but in a manner different from that of AMT. Notably, oral administration of salinomycin together with the neuraminidase inhibitor oseltamivir phosphate (OSV-P) led to enhanced antiviral effect over that with either compound used alone in influenza A virus-infected mouse models. These results provide a new paradigm for developing antivirals and their combination therapy that control both host and viral factors.IMPORTANCE Influenza virus is a main cause of viral respiratory infection in humans as well as animals, occasionally with high mortality. Circulation of influenza viruses resistant to the matrix protein 2 (M2) inhibitor, amantadine, is highly prevalent. Moreover, the frequency of detection of viruses resistant to the neuraminidase inhibitors, including oseltamivir phosphate (OSV-P) or zanamivir, is also increasing. These issues highlight the need for discovery of new antiviral agents with different mechanisms. Salinomycin as the monovalent cation-proton antiporter exhibited consistent inhibitory effects against influenza A and B viruses. It plays multifunctional roles by blocking endosomal acidification and by inactivating the proton transport function of M2, the key steps for influenza virus uncoating. Notably, salinomycin resulted in marked therapeutic effects in influenza virus-infected mice when combined with OSV-P, suggesting that its chemical derivatives could be developed as an adjuvant antiviral therapy to treat influenza infections resistant or less sensitive to existing drugs.


Asunto(s)
Virus de la Influenza A/fisiología , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Oseltamivir/administración & dosificación , Piranos/administración & dosificación , Proteínas de la Matriz Viral/metabolismo , Administración Oral , Animales , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ratones , Proteínas de la Nucleocápside , Infecciones por Orthomyxoviridae/virología , Oseltamivir/farmacología , Transporte de Proteínas/efectos de los fármacos , Piranos/farmacología , Proteínas de Unión al ARN/metabolismo , Proteínas del Núcleo Viral/metabolismo , Internalización del Virus
11.
J Ethnopharmacol ; 209: 236-247, 2017 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-28716571

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Yinqiaosan is a classical traditional Chinese medicine formula, which has been used to treat respiratory diseases since ancient China. It consists of nine herbs and among them, Forsythia suspensa (Thunb.) Vahl fruit is one of the major herbal components. Despite the long history of Yinqiaosan, the active compounds and the mechanisms of action of this formula remain elusive. AIM OF THE STUDY: The present study aimed to examine the suppressive effect of Yinqiaosan on influenza virus and to identify the active components in the formula targeting influenza. MATERIALS AND METHODS: Anti-influenza virus effect of Yinqiaosan was assessed by tissue culture infective dose assay, and was also tested in an in vivo mouse model. Active compound from the formula was identified with a bioactivity-guided fractionation scheme. The potential mode of action of the compound was further investigated by identifying the host cell signaling pathways and viral protein production using in vitro cell culture models. RESULTS: Our results showed that forsythoside A from Forsythia suspensa (Thunb.) Vahl fruit, a major herbal component in Yinqiaosan, reduced the viral titers of different influenza virus subtypes in cell cultures and increased the survival rate of the mice in an in vivo influenza virus infection model. Further experiments on the mode of action of forsythoside A showed that it reduced the influenza M1 protein, which in turn intervened the budding process of the newly formed virions and eventually limited the virus spread. CONCLUSION: Results of our present study provides scientific evidence to support to the application of a traditional herbal formula. We also identify novel candidate compound for future drug development against influenza virus.


Asunto(s)
Forsythia/química , Frutas/química , Glicósidos/farmacología , Virus de la Influenza A/efectos de los fármacos , Infecciones por Orthomyxoviridae/virología , Proteínas de la Matriz Viral/metabolismo , Animales , Antivirales/química , Antivirales/uso terapéutico , Línea Celular , Perros , Relación Dosis-Respuesta a Droga , Regulación Viral de la Expresión Génica/efectos de los fármacos , Glicósidos/administración & dosificación , Glicósidos/química , Ratones , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Proteínas de la Matriz Viral/genética , Cultivo de Virus
12.
Interdiscip Sci ; 9(2): 254-277, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26857866

RESUMEN

Ebola is a deadly virus that has recently emerged as an enormous public health concern which causes dangerous illness with high fatality rates of 90 %. The virus is not receptive to known antivirals, and hence, there is a promising need to identify novel inhibitors to combat the disease. The present study deals with identification of potential herbal leads that probably subdue the activity of four major drug targets of Ebola virus such as VP24, VP30, VP35 and VP40 by computer-aided virtual screening. The selection of receptors was performed based on their functional roles in the disease. The drug likeliness and ADMET parameters of 150 herbal ligands were computationally predicted. Those molecules that qualified these parameters were preferred for docking studies with the protein targets. An existing chemical antiviral drug, BCX4430 was also docked and its theoretical binding energy was scrutinized. The docking studies suggested that herbal ligand Limonin demonstrated high binding properties with VP24 and VP35 (binding energy -9.7 kcal/mol). Similarly, curcumin exhibited good binding with VP30 (binding energy -9.6 kcal/mol). Further, Mahanine displayed superior interaction with VP40 (binding energy -7.7 kcal/mol). These herbal leads demonstrated better binding potential than the known chemical analogue in the computational studies. This study serves to bestow paramount information for further experimental studies concerning the utility of herbal ligands as probable lead molecules against Ebola viral targets.


Asunto(s)
Antivirales/farmacología , Ebolavirus/efectos de los fármacos , Ebolavirus/metabolismo , Adenina/análogos & derivados , Adenosina/análogos & derivados , Descubrimiento de Drogas , Humanos , Simulación del Acoplamiento Molecular , Nucleósidos de Purina/farmacología , Pirrolidinas , Factores de Transcripción/metabolismo , Proteínas de la Matriz Viral/metabolismo , Proteínas Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo
13.
BMC Complement Altern Med ; 14: 284, 2014 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-25088288

RESUMEN

BACKGROUND: We have previously shown that curcumin exhibited an outstanding anti-HCMV effect in vitro and in vivo. However, the underlying mechanism for the anti-HCMV effect remains unclear. METHODS: Levels of IL-6 and TNF-α cytokine secretions in HELF cells were determined by enzyme-linked immunosorbent assay (ELISA); cell cycles were assessed by flow cytometry; ie and ul83 gene expressions were evaluated using reverse transcriptase real-time quantitative PCR; HCMV IE and UL83 antigen expressions were studied using immunofluorescence staining assay and western blot. RESULTS: Curcumin reduced HCMV immediate early antigen (IEA) and UL83A expressions and IL-6, and TNF-α secretions and recovered cell proliferation to normal level in HCMV infected HELF cells. CONCLUSIONS: Curcumin anti-HCMV effect may possibly be that curcumin concurrently alters host cell microenviroment and inhibits the HCMV antigen expressions. These findings may provide a basic understanding of the curcumin anti-HCMV effect and a novel strategy for further development of curcumin anti-HCMV treatment.


Asunto(s)
Curcumina/farmacología , Citomegalovirus/efectos de los fármacos , Línea Celular , Citocinas/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo
14.
Eur J Nutr ; 52(4): 1405-15, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23015061

RESUMEN

PURPOSE: Influenza viruses infect airway epithelial cells, causing respiratory distress. Immune defense is maintained by chemokine/cytokine secretions from airway epithelial cells. While moderate inflammatory response protects from ill effects, hyper-inflammatory response promotes the pathogenesis. High circulating levels of vitamin D are known to mitigate effects of infectious diseases, including respiratory infectious diseases. The question whether and how vitamin D treatment pre-/post-viral exposure modulates inflammatory response is not clear. The present study was undertaken to understand autophagy/apoptosis balance and chemokine/cytokine response to influenza A (H1N1) infection by pre- and post-1, 25-dihydroxyvitamin D3 (1,25[OH]2 D3)[calcitriol] treatment of human lung A549 epithelial cells. METHODS: Influenza A (H1N1) virus was propagated in A549 cell line, titrated using hemagglutination assay, and was used to assess effect of calcitriol. After confirming that 100 nM of calcitriol fails to clear virus, A549 cells were either pre-treated (16 h) with 100 nM or post-treated with 30 nM of 1,25[OH]2 D3 of virus inoculation (1 h). Cells after incubation at 37 °C under 5 % CO2 for 48 h were collected and subjected to RNA and protein extraction. Measurements of viability, influenza M protein, and molecular parameters of cell death and inflammatory response were performed. RESULTS: We report that treatment of these cells with 100/30 nM of 1,25[OH]2 D3 prior to/or post-H1N1 exposure does not affect viral clearance but significantly reduces autophagy and restores increased apoptosis seen on H1N1 infection back to its constitutive level. However, it significantly decreases the levels of H1N1-induced TNF-α (tumor necrosis factor-alpha), IFN-ß (interferon-beta), and IFN-stimulated gene-15 (ISG15). 1,25[OH]2 D3 treatment prior to/or post-H1N1 infection significantly down-regulates IL-8 as well as IL-6 RNA levels. These results demonstrate that calcitriol treatment suppresses the H1N1-induced transcription of the chemokines RANTES and IL-8 in epithelial cells. CONCLUSION: The findings provide support for the initiation of vitamin D supplementation program to VDD populations in reducing the severity of influenza.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Calcitriol/metabolismo , Regulación hacia Abajo , Factores Inmunológicos/metabolismo , Subtipo H1N1 del Virus de la Influenza A/inmunología , Células Epiteliales Alveolares/citología , Células Epiteliales Alveolares/inmunología , Células Epiteliales Alveolares/virología , Apoptosis , Autofagia , Western Blotting , Línea Celular , Supervivencia Celular , Quimiocinas/genética , Quimiocinas/metabolismo , Medios de Cultivo Condicionados/química , Citocinas/genética , Citocinas/metabolismo , Pruebas de Hemaglutinación , Humanos , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Cinética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de la Matriz Viral/metabolismo
15.
Yao Xue Xue Bao ; 46(6): 650-5, 2011 Jun.
Artículo en Chino | MEDLINE | ID: mdl-21882524

RESUMEN

This study is to investigate the treatment of YinQiaojiedu soft capsule for influenza virus A/PR8/34 (H1N1) infection. The model of pneumonia was established by dropping influenza virus into the nose of normal mice, and the lung index and death rate were observed. Real time RT-PCR and Western blotting technique were used to detect the virus load and the relative expression of M1 protein in lungs of mice on the 1st, 3rd, 5th and 7th day after infection. The results showed that YinQiaojiedu soft capsule in 1 g x kg(-1) and 0.5 g x kg(-1) dose groups can decrease the lung index significantly on the 3rd, 5th and 7th day after being infected (P < 0.05, P < 0.01), and the number of death in the two groups of animals decreased significantly. YinQiaojiedu soft capsule in 1 g x kg(-1) dose group can decreased virus load at each time point, and lower it in 0.5 g x kg(-1) dose group at the 3rd, 5th and 7th day (P < 0.05, P < 0.01). YinQiaojiedu soft capsule can decrease the relative expression of M1 protein in lungs of mice, 1 g x kg(-1) and 0.5 g x kg(-1) dose groups are significantly lower in expression of M1 protein compared with model group at the 3rd and 7th day (P < 0.05, P < 0.01). It can be concluded that YinQiaojiedu soft capsule exerts antiviral effects against influenza virus by downregulating expression of virus load and M1 protein.


Asunto(s)
Antivirales/farmacología , Medicamentos Herbarios Chinos/farmacología , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Neumonía/metabolismo , Carga Viral/efectos de los fármacos , Proteínas de la Matriz Viral/metabolismo , Animales , Antivirales/administración & dosificación , Cápsulas , Medicamentos Herbarios Chinos/administración & dosificación , Femenino , Subtipo H1N1 del Virus de la Influenza A , Pulmón/metabolismo , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos ICR , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Neumonía/virología
16.
Blood ; 117(18): 4852-4, 2011 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-21406721

RESUMEN

A20, a negative regulator of NF-κB, has been implicated as a tumor suppressor gene in multiple types of B-cell lymphoma. AIDS-related lymphomas (ARLs) are high-grade B-cell lymphomas that are frequently associated with EBV infection. We examined a panel of ARLs for A20 alterations. FISH showed A20 deletion in 6 of 33 cases (18%). A20 mutations were found in 3 of 19 cases (16%), including 2 cases with deletions of the comple-mentary allele. Immunohistochemistry showed the absence of A20 protein in 7 of 55 samples (13%). In contrast to reports in Hodgkin lymphoma in which EBV infection and A20 alteration are mutually exclusive, A20 inactivation was observed in both EBV(+) and EBV(-) cases. The EBV latent membrane protein 1, which activates NF-κB, was not expressed in 12 of 13 cases with A20 loss. In ARLs loss of A20 may be an alternative mechanism of NF-κB activation in the absence of latent membrane protein 1 expression.


Asunto(s)
Infecciones por Virus de Epstein-Barr/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Linfoma Relacionado con SIDA/genética , Linfoma Relacionado con SIDA/virología , Mutación , Proteínas Nucleares/genética , Análisis Mutacional de ADN , Proteínas de Unión al ADN , Infecciones por Virus de Epstein-Barr/metabolismo , Eliminación de Gen , Silenciador del Gen , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Linfoma Relacionado con SIDA/metabolismo , FN-kappa B/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Proteínas de la Matriz Viral/metabolismo
17.
PLoS One ; 5(2): e9174, 2010 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-20161802

RESUMEN

Recombinant viruses labelled with fluorescent proteins are useful tools in molecular virology with multiple applications (e.g., studies on intracellular trafficking, protein localization, or gene activity). We generated by homologous recombination three recombinant cytomegaloviruses carrying the enhanced yellow fluorescent protein (EYFP) fused with the viral proteins IE-2, ppUL32 (pp150), and ppUL83 (pp65). In growth kinetics, the three viruses behaved all like wild type, even at low multiplicity of infection (MOI). The expression of all three fusion proteins was detected, and their respective localizations were the same as for the unmodified proteins in wild-type virus-infected cells. We established the in vivo measurement of fluorescence intensity and used the recombinant viruses to measure inhibition of viral replication by neutralizing antibodies or antiviral substances. The use of these viruses in a pilot screen based on fluorescence intensity and high-content analysis identified cellular kinase inhibitors that block viral replication. In summary, these viruses with individually EYFP-tagged proteins will be useful to study antiviral substances and the dynamics of viral infection in cell culture.


Asunto(s)
Citomegalovirus/metabolismo , Proteínas Luminiscentes/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Virales/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Antivirales/farmacología , Núcleo Celular/metabolismo , Células Cultivadas , Citomegalovirus/efectos de los fármacos , Citomegalovirus/genética , Evaluación Preclínica de Medicamentos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/virología , Ganciclovir/farmacología , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Luminiscentes/genética , Masculino , Microscopía Fluorescente , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Proteínas Recombinantes de Fusión/genética , Espectrometría de Fluorescencia , Transactivadores/genética , Transactivadores/metabolismo , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo , Proteínas Virales/genética , Replicación Viral/efectos de los fármacos
18.
Protein Pept Lett ; 15(9): 922-30, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18991768

RESUMEN

Influenza A virus matrix M1 protein is membrane associated and plays a crucial role in virus assembly and budding. The N-terminal two thirds of M1 protein was resolved by X-ray crystallography. The overall 3D structure as well as arrangement of the molecule in relation to the viral membrane remains obscure. Now a proteolytic digestion of virions with bromelain was used as an instrument for the in situ assessment of the M1 protein structure. The lipid bilayer around the subviral particles lacking glycoprotein spikes was partially disrupted as was shown by transmission electron microscopy. A phenomenon of M1 protein fragmentation inside the subviral particles was revealed by SDS-PAGE analysis followed by in-gel trypsin hydrolysis and MALDI-TOF mass spectrometry analysis of the additional bands. Putative bromelain-digestion sites appeared to be located at the surface of the M1 protein globule and could be used as landmarks for 3D molecular modeling.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/química , Subtipo H3N2 del Virus de la Influenza A/química , Proteínas de la Matriz Viral/química , Virión/química , Secuencia de Aminoácidos , Bromelaínas/metabolismo , Cristalografía por Rayos X , Electroforesis en Gel de Poliacrilamida , Glicoproteínas Hemaglutininas del Virus de la Influenza , Hidrólisis , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Neuraminidasa , Conformación Proteica , Análisis de Secuencia de Proteína , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Tripsina , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo , Virión/metabolismo
19.
Bioorg Khim ; 34(3): 409-15, 2008.
Artículo en Ruso | MEDLINE | ID: mdl-18672693

RESUMEN

The proteolysis of flu virions of the strain A/Puerto Rico/8/34 (subtype H1N1) by enzymes of various classes was studied to develop an approach to the study of the structural organization and interaction of the basic protein components of the virion environment: hemagglutinin (HA), transmembrane homotrimeric glycoprotein, and matrix protein M1 forming a layer under the lipid membrane. Among the tested proteolytic enzymes and enzymic preparations (thermolysin, trypsin, chymotrypsin, subtilisin Carlsberg, pronase, papain, and bromelain), the cysteine proteases bromelain and papain and the enzymic preparation pronase efficiently deleted HA ectodomains, while chymotrypsin, trypsin, and subtilisin Carlsberg deleted only a part of them. An analysis by MALDI TOF mass spectrometry allowed us to locate the sites of HA hydrolysis by various enzymic preparations. Bromelain, papain, trypsin, and pronase split the polypeptide chain after the K177 residue located before the transmembrane domain (HA2 185-211). Subtilisin Carlsberg hydrolyzed the peptide bond at other neighboring points: after L178 (a basic site) or V176. The hydrolytic activity of bromelain measured by a highly specific chromogenic substrate of cysteine proteases Glp-Phe-Ala-pNA was almost three times higher in the presence of 5 mM beta-mercaptoethanol than in the presence of 50 mM. However, the complete removal of exodomains of HA, HA, and low-activity enzyme by the HA high- and low-activity enzyme required identical time intervals. In the absence of the reducing reagent, the removal of HA by bromelain proceeded a little more slowly and was accompanied by significant fragmentation of protein Ml1. The action of trans-epoxysuccinyl-L-leucylamido)butane (E-64), a specific inhibitor of cysteine proteases, and HgCl2 on the hydrolysis of proteins HA and M1 by bromelain was investigated.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/metabolismo , Péptido Hidrolasas/metabolismo , Virión/metabolismo , Secuencia de Aminoácidos , Bromelaínas/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Hidrólisis , Leucina/análogos & derivados , Leucina/farmacología , Cloruro de Mercurio/farmacología , Datos de Secuencia Molecular , Conformación Proteica , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Proteínas de la Matriz Viral/metabolismo
20.
Emerg Infect Dis ; 13(3): 426-35, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17552096

RESUMEN

Changes in influenza viruses require regular reformulation of strain-specific influenza vaccines. Vaccines based on conserved antigens provide broader protection. Influenza matrix protein 2 (M2) is highly conserved across influenza A subtypes. To evaluate its efficacy as a vaccine candidate, we vaccinated mice with M2 peptide of a widely shared consensus sequence. This vaccination induced antibodies that cross-reacted with divergent M2 peptide from an H5N1 subtype. A DNA vaccine expressing full-length consensus-sequence M2 (M2-DNA) induced M2-specific antibody responses and protected against challenge with lethal influenza. Mice primed with M2-DNA and then boosted with recombinant adenovirus expressing M2 (M2-Ad) had enhanced antibody responses that crossreacted with human and avian M2 sequences and produced T-cell responses. This M2 prime-boost vaccination conferred broad protection against challenge with lethal influenza A, including an H5N1 strain. Vaccination with M2, with key sequences represented, may provide broad protection against influenza A.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Canales Iónicos/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Vacunación , Proteínas de la Matriz Viral/inmunología , Adenoviridae/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Reacciones Cruzadas/inmunología , Evaluación Preclínica de Medicamentos , Femenino , Genes Virales , Vectores Genéticos/administración & dosificación , Vectores Genéticos/metabolismo , Esquemas de Inmunización , Vacunas contra la Influenza/inmunología , Inyecciones Intramusculares , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Infecciones por Orthomyxoviridae/sangre , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/inmunología , Alineación de Secuencia , Linfocitos T/inmunología , Vacunas de ADN/inmunología , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA