Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Curr Opin Virol ; 51: 34-47, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34592709

RESUMEN

The Paramyxoviridae family includes enveloped single-stranded negative-sense RNA viruses such as measles, mumps, human parainfluenza, canine distemper, Hendra, and Nipah viruses, which cause a tremendous global health burden. The ability of paramyxoviral glycoproteins to merge viral and host membranes allows entry of the viral genome into host cells, as well as cell-cell fusion, an important contributor to disease progression. Recent molecular and structural advances in our understanding of the paramyxovirus membrane fusion machinery gave rise to various therapeutic approaches aiming at inhibiting viral infection, spread, and cytopathic effects. These therapeutic approaches include peptide mimics, antibodies, and small molecule inhibitors with various levels of success at inhibiting viral entry, increasing the potential of effective antiviral therapeutic development.


Asunto(s)
Antivirales/farmacología , Membrana Celular/metabolismo , Fusión de Membrana/efectos de los fármacos , Infecciones por Paramyxoviridae/tratamiento farmacológico , Infecciones por Paramyxoviridae/virología , Paramyxoviridae/efectos de los fármacos , Paramyxoviridae/metabolismo , Animales , Antivirales/uso terapéutico , Fusión Celular , Membrana Celular/efectos de los fármacos , Humanos
2.
Antiviral Res ; 175: 104712, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31935422

RESUMEN

Nipah virus (NiV) is a highly pathogenic zoonotic paramyxovirus that continues to cause outbreaks in humans characterized by high mortality and significant clinical sequelae in survivors. Currently, no therapeutics are approved for use in humans against NiV infection. Here, we report that 4'-chloromethyl-2'-deoxy-2'-fluorocytidine (ALS-8112) inhibits NiV. ALS-8112 is the parent nucleoside of lumicitabine, which has been evaluated in phase I and II clinical trials to treat pediatric and adult respiratory syncytial virus infection. In this study, we tested ALS-8112 against NiV and other major human respiratory pneumo- and paramyxoviruses in 2 human lung epithelial cell lines, and demonstrated the ability of ALS-8112 to reduce infectious wild-type NiV yield by over 6 orders of magnitude with no apparent cytotoxicity. However, further cytotoxicity testing in primary cells and bone marrow progenitor cells indicated cytotoxicity at higher concentrations of ALS-8112. Our results warrant the evaluation of lumicitabine against NiV infection in relevant animal models.


Asunto(s)
Antivirales/farmacología , Desoxicitidina/análogos & derivados , Virus Nipah/efectos de los fármacos , Antivirales/química , Línea Celular , Supervivencia Celular/efectos de los fármacos , Desoxicitidina/química , Desoxicitidina/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/virología , Células HeLa , Humanos , Pulmón/citología , Nucleósidos/química , Nucleósidos/farmacología , Paramyxoviridae/efectos de los fármacos
3.
J Virol ; 93(6)2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30567988

RESUMEN

The acute antiviral response is mediated by a family of interferon-stimulated genes (ISGs), providing cell-intrinsic immunity. Mutations in genes encoding these proteins are often associated with increased susceptibility to viral infections. One family of ISGs with antiviral function is the interferon-inducible transmembrane proteins (IFITMs), of which IFITM3 has been studied extensively. In contrast, IFITM1 has not been studied in detail. Since IFITM1 can localize to the plasma membrane, we investigated its function with a range of enveloped viruses thought to infect cells by fusion with the plasma membrane. Overexpression of IFITM1 prevented infection by a number of Paramyxoviridae and Pneumoviridae, including respiratory syncytial virus (RSV), mumps virus, and human metapneumovirus (HMPV). IFITM1 also restricted infection with an enveloped DNA virus that can enter via the plasma membrane, herpes simplex virus 1 (HSV-1). To test the importance of plasma membrane localization for IFITM1 function, we identified blocks of amino acids in the conserved intracellular loop (CIL) domain that altered the subcellular localization of the protein and reduced antiviral activity. By screening reported data sets, 12 rare nonsynonymous single nucleotide polymorphisms (SNPs) were identified in human IFITM1, some of which are in the CIL domain. Using an Ifitm1-/- mouse, we show that RSV infection was more severe, thereby extending the range of viruses restricted in vivo by IFITM proteins and suggesting overall that IFITM1 is broadly antiviral and that this antiviral function is associated with cell surface localization.IMPORTANCE Host susceptibility to viral infection is multifactorial, but early control of viruses not previously encountered is predominantly mediated by the interferon-stimulated gene (ISG) family. There are upwards of 300 of these genes, the majority of which do not have a clearly defined function or mechanism of action. The cellular location of these proteins may have an important effect on their function. One ISG located at the plasma membrane is interferon-inducible transmembrane protein 1 (IFITM1). Here we demonstrate that IFITM1 can inhibit infection with a range of viruses that enter via the plasma membrane. Mutant IFITM1 proteins that were unable to localize to the plasma membrane did not restrict viral infection. We also observed for the first time that IFITM1 plays a role in vivo, and Ifitm1-/- mice were more susceptible to viral lung infection. These data contribute to our understanding of how ISGs prevent viral infections.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Membrana Celular/virología , Paramyxoviridae/efectos de los fármacos , Pneumovirinae/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Células A549 , Secuencia de Aminoácidos , Animales , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Células HEK293 , Humanos , Interferones/farmacología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Células Vero
4.
Sci Rep ; 8(1): 10425, 2018 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-29992955

RESUMEN

Paramyxoviridae, a large family of enveloped viruses harboring a nonsegmented negative-sense RNA genome, include important human pathogens as measles, mumps, respiratory syncytial virus (RSV), parainfluenza viruses, and henipaviruses, which cause some of the deadliest emerging zoonoses. There is no effective antiviral chemotherapy for most of these pathogens. Paramyxoviruses evolved a sophisticated membrane-fusion machine consisting of receptor-binding proteins and the fusion F-protein, critical for virus infectivity. Herein we identify the antiprotozoal/antimicrobial nitazoxanide as a potential anti-paramyxovirus drug targeting the F-protein. We show that nitazoxanide and its circulating-metabolite tizoxanide act at post-entry level by provoking Sendai virus and RSV F-protein aggregate formation, halting F-trafficking to the host plasma membrane. F-protein folding depends on ER-resident glycoprotein-specific thiol-oxidoreductase ERp57 for correct disulfide-bond architecture. We found that tizoxanide behaves as an ERp57 non-competitive inhibitor; the putative drug binding-site was located at the ERp57-b/b' non-catalytic domains interface. ERp57-silencing mimicked thiazolide-induced F-protein alterations, suggesting an important role of this foldase in thiazolides anti-paramyxovirus activity. Nitazoxanide is used in the clinic as a safe and effective antiprotozoal/antimicrobial drug; its antiviral activity was shown in patients infected with hepatitis-C virus, rotavirus and influenza viruses. Our results now suggest that nitazoxanide may be effective also against paramyxovirus infection.


Asunto(s)
Infecciones por Paramyxoviridae/tratamiento farmacológico , Paramyxoviridae/fisiología , Tiazoles/farmacología , Replicación Viral/efectos de los fármacos , Células A549 , Animales , Sitios de Unión , Humanos , Nitrocompuestos , Oxidorreductasas/metabolismo , Paramyxoviridae/efectos de los fármacos , Infecciones por Paramyxoviridae/prevención & control , Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Proteína Disulfuro Isomerasas/química , Pliegue de Proteína/efectos de los fármacos , Transporte de Proteínas , Tiazoles/metabolismo , Proteínas Virales de Fusión/metabolismo
5.
BMC Vet Res ; 14(1): 148, 2018 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-29716604

RESUMEN

BACKGROUND: The aim of the study was to evaluate the impact of herbal extracts on selected immunity mechanisms in clinically healthy pigeons and pigeons inoculated with the pigeon paramyxovirus type 1 (PPMV-1). For the first 7 days post-inoculation (dpi), an aqueous solution of Aloe vera or licorice extract was administered daily at 300 or 500 mg/kg body weight (BW). The birds were euthanized at 4, 7 and 14 dpi, and spleen samples were collected during necropsy. Mononuclear cells were isolated from spleen samples and divided into two parts: one part was used to determine the percentage of IgM+ B cells in a flow cytometric analysis, and the other was used to evaluate the expression of genes encoding IFN-γ and surface receptors on CD3+, CD4+ and CD8+ T cells. RESULTS: The expression of the IFN-γ gene increased in all birds inoculated with PPMV-1 and receiving both herbal extracts. The expression of the CD3 gene was lowest at 14 dpi in healthy birds and at 7 dpi in inoculated pigeons. The expression of the CD4 gene was higher in uninoculated pigeons receiving both herbal extracts than in the control group throughout nearly the entire experiment with a peak at 7 dpi. A reverse trend was observed in pigeons inoculated with PPMV-1 and receiving both herbal extracts. In uninoculated birds, increased expression of the CD8 gene was noted in the pigeons receiving a lower dose of the Aloe vera extract and both doses of licorice extracts. No significant differences in the expression of this gene were found between inoculated pigeons receiving both herbal extracts. The percentage of IgM+ B cells did not differ between any of the evaluated groups. CONCLUSIONS: This results indicate that Aloe vera and licorice extracts have immunomodulatory properties and can be used successfully to prevent viral diseases, enhance immunity and as supplementary treatment for viral diseases in pigeons.


Asunto(s)
Aloe/química , Enfermedades de las Aves/virología , Glycyrrhiza/química , Infecciones por Paramyxoviridae/veterinaria , Paramyxoviridae/efectos de los fármacos , Extractos Vegetales/uso terapéutico , Animales , Enfermedades de las Aves/tratamiento farmacológico , Enfermedades de las Aves/inmunología , Columbidae/inmunología , Columbidae/virología , Citometría de Flujo/veterinaria , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Interferón gamma/metabolismo , Infecciones por Paramyxoviridae/tratamiento farmacológico , Infecciones por Paramyxoviridae/inmunología , Bazo/citología , Bazo/efectos de los fármacos , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología
6.
Antiviral Res ; 144: 147-152, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28629988

RESUMEN

The henipaviruses Nipah virus and Hendra virus are highly pathogenic zoonotic paramyxoviruses which have caused fatal outbreaks of encephalitis and respiratory disease in humans. Despite the availability of a licensed equine Hendra virus vaccine and a neutralizing monoclonal antibody shown to be efficacious against henipavirus infections in non-human primates, there remains no approved therapeutics or vaccines for human use. To explore the possibility of developing small-molecule nucleoside inhibitors against henipaviruses, we evaluated the antiviral activity of 4'-azidocytidine (R1479), a drug previously identified to inhibit flaviviruses, against henipaviruses along with other representative members of the family Paramyxoviridae. We observed similar levels of R1479 antiviral activity across the family, regardless of virus genus. Our brief study expands the documented range of viruses susceptible to R1479, and provides the basis for future investigation and development of 4'-modified nucleoside analogs as potential broad-spectrum antiviral therapeutics across both positive and negative-sense RNA virus families.


Asunto(s)
Antivirales/farmacología , Citidina/análogos & derivados , Paramyxoviridae/efectos de los fármacos , Línea Celular , Citidina/farmacología , Humanos , Pruebas de Sensibilidad Microbiana
7.
Sci Rep ; 7: 43395, 2017 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-28262699

RESUMEN

GS-5734 is a monophosphate prodrug of an adenosine nucleoside analog that showed therapeutic efficacy in a non-human primate model of Ebola virus infection. It has been administered under compassionate use to two Ebola patients, both of whom survived, and is currently in Phase 2 clinical development for treatment of Ebola virus disease. Here we report the antiviral activities of GS-5734 and the parent nucleoside analog across multiple virus families, providing evidence to support new indications for this compound against human viruses of significant public health concern.


Asunto(s)
Alanina/análogos & derivados , Antivirales/farmacología , Ebolavirus/efectos de los fármacos , Marburgvirus/efectos de los fármacos , Paramyxoviridae/efectos de los fármacos , Pneumovirinae/efectos de los fármacos , Profármacos/farmacología , Ribonucleótidos/farmacología , Células A549 , Adenosina Monofosfato/análogos & derivados , Alanina/síntesis química , Alanina/metabolismo , Alanina/farmacología , Animales , Antivirales/síntesis química , Antivirales/metabolismo , Línea Celular Tumoral , Chlorocebus aethiops , Ebolavirus/enzimología , Ebolavirus/crecimiento & desarrollo , Expresión Génica , Células HEK293 , Células HeLa , Hepatocitos/efectos de los fármacos , Hepatocitos/virología , Humanos , Marburgvirus/enzimología , Marburgvirus/crecimiento & desarrollo , Pruebas de Sensibilidad Microbiana , Nucleósidos/síntesis química , Nucleósidos/metabolismo , Nucleósidos/farmacología , Paramyxoviridae/enzimología , Paramyxoviridae/crecimiento & desarrollo , Pneumovirinae/enzimología , Pneumovirinae/crecimiento & desarrollo , Profármacos/síntesis química , Profármacos/metabolismo , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Ribonucleótidos/síntesis química , Ribonucleótidos/metabolismo , Células Vero , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/metabolismo
8.
Sci Rep ; 7: 43610, 2017 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-28344321

RESUMEN

Human paramyxoviruses include global causes of lower respiratory disease like the parainfluenza viruses, as well as agents of lethal encephalitis like Nipah virus. Infection is initiated by viral glycoprotein-mediated fusion between viral and host cell membranes. Paramyxovirus viral fusion proteins (F) insert into the target cell membrane, and form a transient intermediate that pulls the viral and cell membranes together as two heptad-repeat regions refold to form a six-helix bundle structure that can be specifically targeted by fusion-inhibitory peptides. Antiviral potency can be improved by sequence modification and lipid conjugation, and by adding linkers between the protein and lipid components. We exploit the uniquely broad spectrum antiviral activity of a parainfluenza F-derived peptide sequence that inhibits both parainfluenza and Nipah viruses, to investigate the influence of peptide orientation and intervening linker length on the peptides' interaction with transitional states of F, solubility, membrane insertion kinetics, and protease sensitivity. We assessed the impact of these features on biodistribution and antiviral efficacy in vitro and in vivo. The engineering approach based on biophysical parameters resulted in a peptide that is a highly effective inhibitor of both paramyxoviruses and a set of criteria to be used for engineering broad spectrum antivirals for emerging paramyxoviruses.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Paramyxoviridae/efectos de los fármacos , Péptidos/química , Péptidos/farmacología , Proteínas Virales de Fusión/antagonistas & inhibidores , Proteínas Virales de Fusión/química , Secuencia de Aminoácidos , Animales , Antivirales/farmacocinética , Membrana Celular/química , Membrana Celular/metabolismo , Células Cultivadas , Cricetinae , Estructura Molecular , Péptidos/farmacocinética , Unión Proteica , Ratas , Solubilidad , Ensayo de Placa Viral
9.
Eur J Med Chem ; 126: 154-159, 2017 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-27750149

RESUMEN

Thiazolides are polypharmacology agents with at least three mechanisms of action against a broad spectrum of parasites, bacteria and viruses. In respiratory viruses they inhibit the replication of orthomyxoviridae and paramyxoviridae at a post-translational level. Nitazoxanide 1a, the prototype thiazolide, was originally developed as an antiparasitic agent and later repurposed for the treatment of viral respiratory infections. The second generation thiazolides following nitazoxanide, such as the 5-chloro analogue RM-5038 2a, are also broad-spectrum antiviral agents as we have reported. Both 1a and its effective circulating metabolite, tizoxanide 1b, are 5-nitrothiazole derivatives, while RM-5038 2a and its de-acetyl derivative RM-4848 2b are the corresponding 5-chloro derivatives. Recently 1a has completed phase II-III clinical trials in the United States, Canada, Australia and New Zealand in a total of 2865 adults and adolescents of at least 12 months of age with viral acute respiratory illness. Since its biodisposition is primarily seen in the gastro-intestinal tract, its efficacy in systemic viral diseases requires relatively high oral doses. The chemical synthesis of new derivatives with a better systemic absorption was therefore urgently needed. In order to improve their systemic absorption, new amino-ester prodrug derivatives of 1b and RM4848 2b were prepared and tested for their animal pharmacology, pharmacokinetics and toxicology. RM-5061 8a in rats showed 7-fold higher blood concentration compared to 1a: absolute bioavailability increased from 3 to 20%, with a good safety profile in animal safety pharmacology and toxicology.


Asunto(s)
Aminoácidos/química , Antivirales/química , Antivirales/farmacología , Ésteres/química , Profármacos/metabolismo , Tiazoles/síntesis química , Tiazoles/farmacología , Animales , Antivirales/metabolismo , Antivirales/toxicidad , Disponibilidad Biológica , Técnicas de Química Sintética , Evaluación Preclínica de Medicamentos , Femenino , Masculino , Orthomyxoviridae/efectos de los fármacos , Orthomyxoviridae/fisiología , Paramyxoviridae/efectos de los fármacos , Paramyxoviridae/fisiología , Ratas , Seguridad , Tiazoles/metabolismo , Tiazoles/toxicidad , Replicación Viral/efectos de los fármacos
10.
J Virol ; 88(19): 11199-214, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25031353

RESUMEN

Neurotropic alphaviruses, including western, eastern, and Venezuelan equine encephalitis viruses, cause serious and potentially fatal central nervous system infections in humans for which no currently approved therapies exist. We previously identified a series of thieno[3,2-b]pyrrole derivatives as novel inhibitors of neurotropic alphavirus replication, using a cell-based phenotypic assay (W. Peng et al., J. Infect. Dis. 199:950-957, 2009, doi:http://dx.doi.org/10.1086/597275), and subsequently developed second- and third-generation indole-2-carboxamide derivatives with improved potency, solubility, and metabolic stability (J. A. Sindac et al., J. Med. Chem. 55:3535-3545, 2012, doi:http://dx.doi.org/10.1021/jm300214e; J. A. Sindac et al., J. Med. Chem. 56:9222-9241, 2013, http://dx.doi.org/10.1021/jm401330r). In this report, we describe the antiviral activity of the most promising third-generation lead compound, CCG205432, and closely related analogs CCG206381 and CCG209023. These compounds have half-maximal inhibitory concentrations of ∼1 µM and selectivity indices of >100 in cell-based assays using western equine encephalitis virus replicons. Furthermore, CCG205432 retains similar potency against fully infectious virus in cultured human neuronal cells. These compounds show broad inhibitory activity against a range of RNA viruses in culture, including members of the Togaviridae, Bunyaviridae, Picornaviridae, and Paramyxoviridae families. Although their exact molecular target remains unknown, mechanism-of-action studies reveal that these novel indole-based compounds target a host factor that modulates cap-dependent translation. Finally, we demonstrate that both CCG205432 and CCG209023 dampen clinical disease severity and enhance survival of mice given a lethal western equine encephalitis virus challenge. These studies demonstrate that indole-2-carboxamide compounds are viable candidates for continued preclinical development as inhibitors of neurotropic alphaviruses and, potentially, of other RNA viruses. IMPORTANCE There are currently no approved drugs to treat infections with alphaviruses. We previously identified a novel series of compounds with activity against these potentially devastating pathogens (J. A. Sindac et al., J. Med. Chem. 55:3535-3545, 2012, doi:http://dx.doi.org/10.1021/jm300214e; W. Peng et al., J. Infect. Dis. 199:950-957, 2009, doi:http://dx.doi.org/10.1086/597275; J. A. Sindac et al., J. Med. Chem. 56:9222-9241, 2013, http://dx.doi.org/10.1021/jm401330r). We have now produced third-generation compounds with enhanced potency, and this manuscript provides detailed information on the antiviral activity of these advanced-generation compounds, including activity in an animal model. The results of this study represent a notable achievement in the continued development of this novel class of antiviral inhibitors.


Asunto(s)
Antivirales/farmacología , Virus de la Encefalitis Equina del Oeste/efectos de los fármacos , Encefalomielitis Equina/tratamiento farmacológico , Indoles/farmacología , Piridinas/farmacología , Replicación Viral/efectos de los fármacos , Animales , Antivirales/síntesis química , Bunyaviridae/efectos de los fármacos , Bunyaviridae/crecimiento & desarrollo , Línea Celular , Virus de la Encefalitis Equina del Oeste/crecimiento & desarrollo , Virus de la Encefalitis Equina del Oeste/patogenicidad , Encefalomielitis Equina/mortalidad , Encefalomielitis Equina/virología , Femenino , Indoles/síntesis química , Concentración 50 Inhibidora , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/virología , Paramyxoviridae/efectos de los fármacos , Paramyxoviridae/crecimiento & desarrollo , Picornaviridae/efectos de los fármacos , Picornaviridae/crecimiento & desarrollo , Biosíntesis de Proteínas/efectos de los fármacos , Piridinas/síntesis química , Replicón/efectos de los fármacos , Relación Estructura-Actividad , Análisis de Supervivencia
11.
Viruses ; 6(7): 2531-50, 2014 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-24967693

RESUMEN

RNA viruses are capable of rapid spread and severe or potentially lethal disease in both animals and humans. The development of reverse genetics systems for manipulation and study of RNA virus genomes has provided platforms for designing and optimizing viral mutants for vaccine development. Here, we review the impact of RNA virus reverse genetics systems on past and current efforts to design effective and safe viral therapeutics and vaccines.


Asunto(s)
Genoma Viral , Infecciones por Virus ARN/prevención & control , Genética Inversa/métodos , Vacunas Virales/genética , Animales , Coronaviridae/efectos de los fármacos , Coronaviridae/genética , Coronaviridae/inmunología , Flaviviridae/efectos de los fármacos , Flaviviridae/genética , Flaviviridae/inmunología , Ingeniería Genética , Humanos , Orthomyxoviridae/efectos de los fármacos , Orthomyxoviridae/genética , Orthomyxoviridae/inmunología , Paramyxoviridae/efectos de los fármacos , Paramyxoviridae/genética , Paramyxoviridae/inmunología , Infecciones por Virus ARN/inmunología , Infecciones por Virus ARN/virología , Vacunas Virales/administración & dosificación , Vacunas Virales/biosíntesis
12.
Nat Prod Commun ; 9(1): 51-4, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24660461

RESUMEN

Wild berry species are known to exhibit a wide range of pharmacological activities. They have long been traditionally applied for their antiseptic, antimicrobial, cardioprotective and antioxidant properties. The aim of the present study is to reveal the potential for selective antiviral activity of total methanol extracts, as well as that of the anthocyanins and the non-anthocyanins from the following wild berries picked in Bulgaria: strawberry (Fragaria vesca L.) and raspberry (Rubus idaeus L.) of the Rosaceae plant family, and bilberry (Vaccinium myrtillis L.) and lingonberry (Vaccinium vitis-idaea L) of the Ericaceae. The antiviral effect has been tested against viruses that are important human pathogens and for which chemotherapy and/or chemoprophylaxis is indicated, namely poliovirus type 1 (PV-1) and coxsackievirus B1 (CV-B1) from the Picornaviridae virus family, human respiratory syncytial virus A2 (HRSV-A2) from the Paramyxoviridae and influenza virus A/H3N2 of Orthomyxoviridae. Wild berry fruits are freeze-dried and ground, then total methanol extracts are prepared. Further the extracts are fractioned by solid phase extraction and the non-anthocyanin and anthocyanin fractions are eluted. The in vitro antiviral effect is examined by the virus cytopathic effect (CPE) inhibition test. The results reveal that the total extracts of all tested berry fruits inhibit the replication of CV-B1 and influenza A virus. CV-B1 is inhibited to the highest degree by both bilberry and strawberry, as well as by lingonberry total extracts, and influenza A by bilberry and strawberry extracts. Anthocyanin fractions of all wild berries strongly inhibit the replication of influenza virus A/H3N2. Given the obtained results it is concluded that wild berry species are a valuable resource of antiviral substances and the present study should serve as a basis for further detailed research on the matter.


Asunto(s)
Antivirales/análisis , Ericaceae/química , Rosaceae/química , Animales , Línea Celular Tumoral , Perros , Frutas/química , Humanos , Células de Riñón Canino Madin Darby , Pruebas de Sensibilidad Microbiana , Orthomyxoviridae/efectos de los fármacos , Paramyxoviridae/efectos de los fármacos , Picornaviridae/efectos de los fármacos , Extractos Vegetales/farmacología
13.
Antimicrob Agents Chemother ; 57(2): 983-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23229488

RESUMEN

Few antiviral agents are available for treating paramyxovirus infections, such as those involving respiratory syncytial virus (RSV), parainfluenza virus (PIV), and human metapneumovirus (hMPV). We evaluated the effect of oral ribavirin on clinical outcomes of paramyxovirus infections in patients with hematological diseases. All adult patients with paramyxovirus were retrospectively reviewed over a 2-year period. Patients who received oral ribavirin were compared to those who received supportive care without ribavirin therapy. A propensity-matched case-control study and a logistic regression model with inverse probability of treatment weighting (IPTW) were performed to reduce the effect of selection bias in assignment for oral ribavirin therapy. A total of 145 patients, including 64 (44%) with PIV, 60 (41%) with RSV, and 21 (15%) with hMPV, were analyzed. Of these 145 patients, 114 (78%) received oral ribavirin and the remaining 31 (21%) constituted the nonribavirin group. Thirty-day mortality and underlying respiratory death rates were 31% (35/114) and 12% (14/114), respectively, for the oral ribavirin group versus 19% (6/31) and 16% (5/31), respectively, for the nonribavirin group (P = 0.21 and P = 0.56). In the case-control study, the 30-day mortality rate in the ribavirin group was 24% (5/21) versus 19% (4/21) in the nonribavirin group (P = 0.71). In addition, the logistic regression model with IPTW revealed no significant difference in 30-day mortality (adjusted hazard ratio of 1.3; 95% confidence interval [95% CI] of 0.3 to 5.8) between the two groups. Steroid use (adjusted odds ratio, 5.67; P = 0.01) and upper respiratory tract infection (adjusted odds ratio, 0.07; P = 0.001) was independently associated with mortality. Our data suggest that oral ribavirin therapy may not improve clinical outcomes in hematologic disease patients infected with paramyxovirus.


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Paramyxoviridae/tratamiento farmacológico , Infecciones por Paramyxoviridae/mortalidad , Ribavirina/uso terapéutico , Adulto , Anciano , Antivirales/administración & dosificación , Estudios de Casos y Controles , Femenino , Enfermedades Hematológicas/virología , Humanos , Masculino , Persona de Mediana Edad , Paramyxoviridae/efectos de los fármacos , Puntaje de Propensión , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Infecciones del Sistema Respiratorio/mortalidad , Estudios Retrospectivos , Ribavirina/administración & dosificación , Resultado del Tratamiento
14.
Antibiot Khimioter ; 55(7-8): 13-6, 2010.
Artículo en Ruso | MEDLINE | ID: mdl-21140558

RESUMEN

The Ingavirin antiviral properties with respect to the parainfluenza virus, as an actual human respiratory tract pathogen, were investigated by two methods, i.e. immunoenzymatic analysis and microtetrazolium test. The results showed that along with the immediate antiviral activity Ingavirin had nonspecific cytoprotective properties. While affecting the virus proteins synthesis, Ingavirin lowered the virus cytopathogenic action. The drug significantly decreased the portion of the bronchial epithelium cells killed at the stage of acute infection.


Asunto(s)
Amidas/farmacología , Antivirales/farmacología , Efecto Citopatogénico Viral/efectos de los fármacos , Citoprotección , Ácidos Dicarboxílicos/farmacología , Imidazoles/farmacología , Paramyxoviridae/efectos de los fármacos , Animales , Bronquios/efectos de los fármacos , Bronquios/patología , Bronquios/virología , Caproatos , Línea Celular , Humanos , Técnicas para Inmunoenzimas , Infecciones por Paramyxoviridae/patología , Infecciones por Paramyxoviridae/virología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología , Sales de Tetrazolio/química
15.
Microbiol Immunol ; 49(9): 835-44, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16172538

RESUMEN

Effects of proteasome inhibitors on the replication of a paramyxovirus in comparison with the effects on replication of an orthomyxovirus and rhabdovirus were investigated. Treatment of Sendai virus (SeV)-infected LLC-MK2 cells with 50 microM MG132 reduced virus growth to ca. 1/10,000, and treatment with different concentrations of MG132 reduced virus growth in a dose-dependent manner. Released amounts of viral proteins were reduced in correspondence with decrease in infectivity. The inhibition of virus maturation was confirmed by an SeV-like particle formation system. Lactacystin also impaired SeV growth and zLL impaired the growth to a lesser extent, suggesting involvement of proteasomes in the restriction of virus growth. In the presence of MG132, localizations of the M protein and viral F and HN glycoproteins on the cell membrane appeared to be partly dissociated, although the viral glycoproteins were normally transported to the cell surface. These results suggest that an early step of SeV assembly was disturbed by proteasome inhibitors. The relationship of the results with ubiquitin is also discussed. SeV maturation was less susceptible and resistant to MG132 in CV1 cells and A549 cells, respectively, indicating cell specificity of the drug effect. Release of vesicular stomatitis virus also showed high susceptibility to MG132 and release of influenza virus A/WSN/33 was only mildly susceptible to the drug in LLC-MK2 cells. Effects of proteasome inhibitors on virus maturation are thus highly cell-specific and partly virus-specific.


Asunto(s)
Leupeptinas/farmacología , Orthomyxoviridae/efectos de los fármacos , Paramyxoviridae/efectos de los fármacos , Inhibidores de Proteasoma , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacología , Animales , Línea Celular , Chlorocebus aethiops , Inhibidores de Cisteína Proteinasa/farmacología , Proteína HN/metabolismo , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/crecimiento & desarrollo , Macaca mulatta , Macrófagos/virología , Orthomyxoviridae/crecimiento & desarrollo , Paramyxoviridae/crecimiento & desarrollo , Rhabdoviridae/efectos de los fármacos , Rhabdoviridae/crecimiento & desarrollo , Virus Sendai/efectos de los fármacos , Virus Sendai/crecimiento & desarrollo , Virus de la Estomatitis Vesicular Indiana/efectos de los fármacos , Virus de la Estomatitis Vesicular Indiana/crecimiento & desarrollo , Proteínas Virales de Fusión/metabolismo , Proteínas de la Matriz Viral/metabolismo , Ensamble de Virus/efectos de los fármacos , Ensamble de Virus/fisiología , Replicación Viral/efectos de los fármacos , Replicación Viral/fisiología
16.
J Virol Methods ; 128(1-2): 29-36, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16023521

RESUMEN

The Paramyxoviridae form a large family of viruses containing many human and veterinary pathogens for which a need for antiviral treatment is emphasized, particularly following the recent emergence of new viruses. The viral RNA-dependent RNA polymerase constitutes an obvious target for antiviral compounds. An in vitro assay was developed that allows high throughput screening of compounds potentially inhibiting the Sendai virus RNA-dependent RNA polymerase. Screening relies on the detection of the Photinus pyralis luciferase produced in a transcription/translation coupled assay using a mini-replicon virus. It contains an internal control for possible adverse effects of the tested compounds on translation or on luciferase activity. It is estimated that the mini-replicon template produced in one fertilized egg is sufficient to run 5000-10,000 reactions. This assay constitutes a simple, sensitive and easily automated method to perform high throughput screening of Paramyxoviridae RNA-dependent RNA polymerase inhibitors.


Asunto(s)
Antivirales/farmacología , Inhibidores Enzimáticos/farmacología , Pruebas de Sensibilidad Microbiana/métodos , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , ARN Polimerasa Dependiente del ARN/metabolismo , Virus Sendai/enzimología , Animales , Luciérnagas/enzimología , Células HeLa , Humanos , Luciferasas/metabolismo , Paramyxoviridae/efectos de los fármacos , Paramyxoviridae/enzimología , Paramyxoviridae/genética , Biosíntesis de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Virus Sendai/efectos de los fármacos , Virus Sendai/genética , Virus Sendai/metabolismo , Transcripción Genética
18.
Z Naturforsch C J Biosci ; 53(9-10): 883-7, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9825543

RESUMEN

Cholesteryl 3",4"-dimethoxycinnamate (7) and a new synthesized o-coumaroyl ester of 3 beta-(2'-hydroxyethoxy)-cholest-5-en (13) exhibited a marked activity against poliovirus type 1 (Mahoney). Compound 7 showed an approximately 20-fold greater selectivity in its antiviral activity than compound 13. These compounds were selected from thirteen steryl esters of cinnamic acid derivatives through an in vitro antiviral screening against viruses belonging to taxonomic groups with causative agents of important human infectious diseases to which chemotherapy is indicated, i.e. Picornaviridae, Orthomyxoviridae, Paramyxoviridae and Herpesviridae.


Asunto(s)
Antivirales/síntesis química , Ésteres del Colesterol/síntesis química , Cinamatos/síntesis química , Antivirales/química , Antivirales/farmacología , Ésteres del Colesterol/química , Ésteres del Colesterol/farmacología , Cinamatos/química , Cinamatos/farmacología , Herpesviridae/efectos de los fármacos , Humanos , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Orthomyxoviridae/efectos de los fármacos , Paramyxoviridae/efectos de los fármacos , Picornaviridae/efectos de los fármacos , Poliovirus/efectos de los fármacos , Relación Estructura-Actividad , Virosis
19.
Tohoku J Exp Med ; 177(4): 315-25, 1995 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8928191

RESUMEN

A rapid and precise screening assay was developed for in vitro evaluation of anti-orthomyxo- and anti-paramyxovirus agents. The procedure is spectrophotometrical assessment for viability of cells via extracellular leakage of lactic dehydrogenase (LDH). HMV-II cells, a human melanoma cell line was found to be suitable for the titration of virus infectivity and screening of anti-viral agents for orthomyxo- and paramyxoviruses. Comparative titration of infectivity of stock viruses by the LDH and the MTT in site reduction of 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) methods with HMV-II cells as well as plaque titration with MDCK, Vero and HeLa cells was carried out. The LDH method was comparable or more sensitive for influenza viruses (FLUV)-A, B, C, parainfluenza viruses (PFLUV)-1, 2 and less sensitive for PFLUV-3, mumps virus (MPSV), measles viruses (MLSV) and respiratory syncytial virus (RSV) than the plaque titration. The 50% effective concentration (EC50) of 1-beta-D-ribofuranosyl-1, 2, 4-triazol-3-carboxamide (ribavirin) and 5-ethynyl-1-beta-D-ribofuranosyl-imidazole-4-carboxamide (EICAR) against orthomyxo- and paramyxoviruses were examined comparatively by the LDH, MTT and plaque reduction (PR) methods. The EC50 values of FLUV-C and PFLUV-1 were able to be evaluated only by the LDH but not by the MTT and PR methods. The LDH method with HMV-II cells simplifies the assay procedure and permits the evaluation of a large number of compounds for anti-orthomyxo- and anti-paramyxoviruses activity in vitro.


Asunto(s)
Antivirales/farmacología , L-Lactato Deshidrogenasa/análisis , Orthomyxoviridae/efectos de los fármacos , Orthomyxoviridae/patogenicidad , Paramyxoviridae/efectos de los fármacos , Paramyxoviridae/patogenicidad , Colorimetría , Efecto Citopatogénico Viral/efectos de los fármacos , Humanos , Melanoma Experimental/enzimología , Ribavirina/farmacología , Ribonucleósidos/farmacología , Sales de Tetrazolio , Tiazoles , Células Tumorales Cultivadas , Ensayo de Placa Viral
20.
Arch Virol ; 130(3-4): 317-26, 1993.
Artículo en Inglés | MEDLINE | ID: mdl-8517791

RESUMEN

The effect of dextran sulfate on the fusion of a series of enveloped viruses, bearing specifically different fusion proteins, was investigated. The fusion with model- and with biological membranes was monitored by an R18 fluorescence-dequenching fusion assay. Dextran sulfate strongly suppresses the fusion of orthomxyo- (influenza A (H1N1 and H3N2 subtypes) and influenza B), of toga- (Semliki Forest virus), and of rhabdoviruses (vesicular stomatitis and rabies virus). The fusion of the paramyxo-viruses Sendai and mumps was not significantly affected by the anionic polysaccharide. The response to dextran sulfate was virus-specific, and identical for the different members of one virusfamily, bearing the same fusion protein. It was shown that dextran sulfate attaches with high affinity to the viruses studied, but not to erythrocytes. The anionic polymer appears to attach to the fusion epitope of the viral membrane. The inhibition of virus replication in vitro shows a remarkable correlation with the observed anti-fusion effects of dextran sulfate.


Asunto(s)
Sulfato de Dextran/farmacología , Fusión de Membrana/efectos de los fármacos , Virus ARN/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Membrana Eritrocítica/microbiología , Humanos , Concentración de Iones de Hidrógeno , Técnicas In Vitro , Liposomas , Orthomyxoviridae/efectos de los fármacos , Paramyxoviridae/efectos de los fármacos , Rhabdoviridae/efectos de los fármacos , Virus de los Bosques Semliki/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA