Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Peptides ; 142: 170570, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34000327

RESUMEN

Viruses remain one of the leading causes of animal and human disease. Some animal viral infections spread sporadically to human populations, posing a serious health risk. Particularly the emerging viral zoonotic diseases such as the novel, zoonotic coronavirus represent an actual challenge for the scientific and medical community. Besides human health risks, some animal viral infections, although still not zoonotic, represent important economic loses to the livestock industry. Viral infections pose a genuine concern for which there has been an increasing interest for new antiviral molecules. Among these novel compounds, antiviral peptides have been proposed as promising therapeutic options, not only for the growing body of evidence showing hopeful results but also due to the many adverse effects of chemical-based drugs. Here we review the current progress, key targets and considerations for the development of antiviral peptides (AVPs). The review summarizes the state of the art of the AVPs tested in zoonotic (coronaviruses, Rift Valley fever viruses, Eastern Equine Encephalitis Virus, Dengue and Junín virus) and also non-zoonotic farm animal viruses (avian and cattle viruses). Their molecular target, amino acid sequence and mechanism of action are summarized and reviewed. Antiviral peptides are currently on the cutting edge since they have been reported to display anti-coronavirus activity. Particularly, the review will discuss the specific mode of action of AVPs that specifically inhibit the fusion of viral and host-cell membranes for SARS-CoV-2, showing in detail some important features of the fusion inhibiting peptides that target the spike protein of these risky viruses.


Asunto(s)
Péptidos/farmacología , Zoonosis Virales/tratamiento farmacológico , Virus/efectos de los fármacos , Animales , Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Virus de la Encefalitis Equina del Este/efectos de los fármacos , Humanos , Virus Junin/efectos de los fármacos , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos
2.
Viruses ; 13(4)2021 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-33805122

RESUMEN

Rift Valley fever virus (RVFV) is a mosquito-borne bunyavirus that causes an important disease in ruminants, with great economic losses. The infection can be also transmitted to humans; therefore, it is considered a major threat to both human and animal health. In a previous work, we described a novel RVFV variant selected in cell culture in the presence of the antiviral agent favipiravir that was highly attenuated in vivo. This variant displayed 24 amino acid substitutions in different viral proteins when compared to its parental viral strain, two of them located in the NSs protein that is known to be the major virulence factor of RVFV. By means of a reverse genetics system, in this work we have analyzed the effect that one of these substitutions, P82L, has in viral attenuation in vivo. Rescued viruses carrying this single amino acid change were clearly attenuated in BALB/c mice while their growth in an interferon (IFN)-competent cell line as well as the production of interferon beta (IFN-ß) did not seem to be affected. However, the pattern of nuclear NSs accumulation was modified in cells infected with the mutant viruses. These results highlight the key role of the NSs protein in the modulation of viral infectivity.


Asunto(s)
Sustitución de Aminoácidos , Fiebre del Valle del Rift/prevención & control , Virus de la Fiebre del Valle del Rift/química , Virus de la Fiebre del Valle del Rift/inmunología , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Amidas/farmacología , Animales , Antivirales/farmacología , Línea Celular , Chlorocebus aethiops , Cricetinae , Células HEK293 , Humanos , Riñón/citología , Masculino , Ratones , Ratones Endogámicos BALB C , Pirazinas/farmacología , Genética Inversa , Fiebre del Valle del Rift/inmunología , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Virus de la Fiebre del Valle del Rift/genética , Células Vero , Virulencia , Factores de Virulencia/genética
3.
Monoclon Antib Immunodiagn Immunother ; 40(2): 60-64, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33900823

RESUMEN

The DNA fragment encoding predicted main antigenic region, aa 14-245 on N protein of Rift Valley virus (RVFV) was cloned into the vector pET-28a (+) and p3xFLAG-CMV-10. The recombinant pET-28a-N1 protein was expressed in Escherichia coli BL21 (DE3) with 1 mM isopropyl-b-thio-galactopyranoside at 37°C for 5 hours, and purified by protein purifier. Three monoclonal antibodies (mAbs) named 3A5, 3A6, and 3A7 against N protein were obtained by fusing mouse myeloma cell line SP2/0 with spleen lymphocytes from pET-28a-N1 protein-immunized mice. Finally, the mAbs were characterized by enzyme-linked immunosorbent assays, indirect immunofluorescent assays, and Western blot. The results show that all the mAbs possess high specificity and react with both prokaryotic and eukaryotic N protein, which could provide important materials for the research on the function of N protein and the diagnostic methods of RVFV.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Proteínas de la Nucleocápside/antagonistas & inhibidores , Fiebre del Valle del Rift/terapia , Virus de la Fiebre del Valle del Rift/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Línea Celular Tumoral , Humanos , Ratones , Proteínas de la Nucleocápside/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Fiebre del Valle del Rift/inmunología , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Virus de la Fiebre del Valle del Rift/patogenicidad
4.
Viruses ; 13(2)2021 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-33572659

RESUMEN

Rift valley fever virus (RVFV) is a mosquito-borne virus endemic to sub-Saharan African countries, and the first sporadic outbreaks outside Africa were reported in the Asia-Pacific region. There are no approved therapeutic agents available for RVFV; however, finding an effective antiviral agent against RVFV is important. This study aimed to evaluate the antiviral, antioxidant and anti-inflammatory activity of medicinal plant extracts. Twenty medicinal plants were screened for their anti-RVFV activity using the cytopathic effect (CPE) reduction method. The cytotoxicity assessment of the extracts was done before antiviral screening using the MTT assay. Antioxidant and reactive oxygen/nitrogen species' (ROS/RNS) inhibitory activity by the extracts was investigated using non-cell-based and cell-based assays. Out of twenty plant extracts tested, eight showed significant potency against RVFV indicated by a decrease in tissue culture infectious dose (TCID50) < 105. The cytotoxicity of extracts showed inhibitory concentrations values (IC50) > 200 µg/mL for most of the extracts. The antioxidant activity and anti-inflammatory results revealed that extracts scavenged free radicals exhibiting an IC50 range of 4.12-20.41 µg/mL and suppressed the production of pro-inflammatory mediators by 60-80% in Vero cells. This study demonstrated the ability of the extracts to lower RVFV viral load and their potency to reduce free radicals.


Asunto(s)
Antiinflamatorios/farmacología , Antioxidantes/farmacología , Antivirales/farmacología , Extractos Vegetales/farmacología , Plantas Medicinales/química , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Animales , Antiinflamatorios/química , Antioxidantes/química , Antivirales/química , Chlorocebus aethiops , Evaluación Preclínica de Medicamentos , Extractos Vegetales/química , Fiebre del Valle del Rift/tratamiento farmacológico , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/crecimiento & desarrollo , Sudáfrica , Células Vero
5.
J Virol Methods ; 276: 113794, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31794780

RESUMEN

Ensuring the successful inactivation of select agent material is critical for maintaining compliance with federal regulations and safeguarding laboratory personnel from exposure to dangerous pathogens. Rift Valley fever virus (RVFV), naturally transmitted by mosquitoes, is classified as a select agent by the CDC and USDA due to its potential to cause significant economic losses to the livestock industry and its demonstrated potential to emerge into naïve geographic areas. Herein we describe several effective inactivation procedures for RVFV infected mosquito samples. We also demonstrate the vaccine strain MP-12 can be used as an appropriate analog for inactivation testing and describe a method of validating inactivation using Amicon filters. Briefly, we show the following inactivation methods are all effective at inactivating RVFV and MP-12 by following the manufacturers'/established protocols: 4 % paraformaldehyde, Trizol LS (ThermoFisher Scientific), MagMAX™-96 Viral RNA Isolation Kit (ThermoFisher Scientific), and Mag-Bind® Viral DNA/RNA 96 Kit (Omega Bio-Tek).


Asunto(s)
Culex/virología , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Virus de la Fiebre del Valle del Rift/fisiología , Virología/métodos , Inactivación de Virus , Animales , Chlorocebus aethiops , Culex/efectos de los fármacos , Células Vero
6.
Artículo en Inglés | MEDLINE | ID: mdl-31085519

RESUMEN

Rift Valley fever virus (RVFV) is an emerging, mosquito-borne, zoonotic pathogen with recurrent outbreaks taking a considerable toll in human deaths in many African countries, for which no effective treatment is available. In cell culture studies and with laboratory animal models, the nucleoside analogue favipiravir (T-705) has demonstrated great potential for the treatment of several seasonal, chronic, and emerging RNA virus infections in humans, suggesting applicability to control some viral outbreaks. Treatment with favipiravir was shown to reduce the infectivity of Rift Valley fever virus both in cell cultures and in experimental animal models, but the mechanism of this protective effect is not understood. In this work, we show that favipiravir at concentrations well below the toxicity threshold estimated for cells is able to extinguish RVFV from infected cell cultures. Nucleotide sequence analysis has documented RVFV mutagenesis associated with virus extinction, with a significant increase in G to A and C to U transition frequencies and a decrease of specific infectivity, hallmarks of lethal mutagenesis.


Asunto(s)
Amidas/farmacología , Mutagénesis/genética , Pirazinas/farmacología , Virus de la Fiebre del Valle del Rift/genética , Animales , Secuencia de Bases , Línea Celular , Chlorocebus aethiops , Culicidae , Mutagénesis/efectos de los fármacos , ARN Viral/genética , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Células Vero
7.
Vet Microbiol ; 230: 110-116, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30827375

RESUMEN

Rift Valley fever virus (RVFV) is the causative agent of Rift Valley fever (RVF) that affects both livestock and humans. There are neither fully licensed RVF vaccines available for human or animal use, nor effective antiviral drugs approved for human use in the U.S. To identify antiviral compounds effective for RVF, we developed and employed a cell-based high-throughput assay using a recombinant RVFV MP-12 strain, which expresses Renilla luciferase in place of the NSs protein, to screen 727 small compounds purchased from the National Institutes of Health. Twenty-three compounds were initially identified using the screening assay. Two compounds, 6-azauridine and mitoxantrone, also inhibited the replication of the parental MP-12 strain encoding the NSs gene, with limited cytotoxic effects. The respective 50% inhibitory concentrations were 29.07 µM and 79.85 µM when tested with the parental MP-12 strain at a multiplicity of infection of 2. The compounds were further evaluated using the STAT-1 KO mouse model. At one hour post intranasal inoculation of MP-12 strain, mice were intranasally treated with each indicated compound twice daily. Mice treated with either placebo or 6-azauridine displayed severe weight loss and reached the threshold for euthanasia with obvious neurologic symptoms. Onset of disease was, however, delayed in mice treated with either ribavirin or mitoxantrone. The results indicated that mitoxantrone can reduce the severity of diseases in RVFV-infected mice. Our studies build the foundation for the initial screening and efficacy studies of RVF antivirals in a BSL-2 environment, avoiding the higher risks of BSL-3 exposure with wild-type virus.


Asunto(s)
Antivirales/farmacología , Fiebre del Valle del Rift/tratamiento farmacológico , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Animales , Antivirales/aislamiento & purificación , Azauridina/farmacología , Línea Celular , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Femenino , Ensayos Analíticos de Alto Rendimiento , Concentración 50 Inhibidora , Ratones , Mitoxantrona/farmacología , Virus de la Fiebre del Valle del Rift/fisiología , Bibliotecas de Moléculas Pequeñas/farmacología , Replicación Viral/efectos de los fármacos
8.
Cell Rep ; 25(13): 3750-3758.e4, 2018 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-30590046

RESUMEN

The Gn subcomponent of the Gn-Gc assembly that envelopes the human and animal pathogen, Rift Valley fever virus (RVFV), is a primary target of the neutralizing antibody response. To better understand the molecular basis for immune recognition, we raised a class of neutralizing monoclonal antibodies (nAbs) against RVFV Gn, which exhibited protective efficacy in a mouse infection model. Structural characterization revealed that these nAbs were directed to the membrane-distal domain of RVFV Gn and likely prevented virus entry into a host cell by blocking fusogenic rearrangements of the Gn-Gc lattice. Genome sequence analysis confirmed that this region of the RVFV Gn-Gc assembly was under selective pressure and constituted a site of vulnerability on the virion surface. These data provide a blueprint for the rational design of immunotherapeutics and vaccines capable of preventing RVFV infection and a model for understanding Ab-mediated neutralization of bunyaviruses more generally.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Antivirales/farmacología , Virus de la Fiebre del Valle del Rift/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Neutralizantes/farmacología , Chlorocebus aethiops , Femenino , Glicoproteínas/química , Glicoproteínas/metabolismo , Células HEK293 , Humanos , Inmunización , Inmunoglobulina G/metabolismo , Ratones Endogámicos BALB C , Modelos Biológicos , Pruebas de Neutralización , Dominios Proteicos , Conejos , Proteínas Recombinantes/farmacología , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Células Vero , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo
9.
Antiviral Res ; 156: 38-45, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29864447

RESUMEN

Rift Valley fever virus (RVFV) is a mosquito-borne pathogen endemic to sub-Saharan Africa and the Arabian Peninsula. There are no approved antiviral therapies or vaccines available to treat or prevent severe disease associated with RVFV infection in humans. The adenosine analog, galidesivir (BCX4430), is a broad-spectrum antiviral drug candidate with in vitro antiviral potency (EC50 of less than 50 µM) in more than 20 different viruses across eight different virus families. Here we report on the activity of galidesivir in the hamster model of peracute RVFV infection. Intramuscular and intraperitoneal treatments effectively limited systemic RVFV (strain ZH501) infection as demonstrated by significantly improved survival outcomes and the absence of infectious virus in the spleen and the majority of the serum, brain, and liver samples collected from infected animals. Our findings support the further development of galidesivir as an antiviral therapy for use in treating severe RVFV infection, and possibly other related phleboviral diseases.


Asunto(s)
Antivirales/administración & dosificación , Nucleósidos de Purina/administración & dosificación , Fiebre del Valle del Rift/tratamiento farmacológico , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Adenina/análogos & derivados , Adenosina/análogos & derivados , Animales , Modelos Animales de Enfermedad , Inyecciones Intramusculares , Inyecciones Intraperitoneales , Hígado/virología , Mesocricetus , Pirrolidinas , Bazo/virología , Análisis de Supervivencia , Resultado del Tratamiento
10.
Viruses ; 10(4)2018 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-29652799

RESUMEN

Viruses must parasitize host cell translational machinery in order to make proteins for viral progeny. In this study, we sought to use this signal transduction conduit against them by inhibiting multiple kinases that influence translation. Previous work indicated that several kinases involved in translation, including p70 S6K, p90RSK, ERK, and p38 MAPK, are phosphorylated following Rift Valley fever virus (RVFV) infection. Furthermore, inhibiting p70 S6K through treatment with the FDA approved drug rapamycin prevents RVFV pathogenesis in a mouse model of infection. We hypothesized that inhibiting either p70 S6K, p90RSK, or p90RSK’s upstream kinases, ERK and p38 MAPK, would decrease translation and subsequent viral replication. Treatment with the p70 S6K inhibitor PF-4708671 resulted in decreased phosphorylation of translational proteins and reduced RVFV titers. In contrast, treatment with the p90RSK inhibitor BI-D1870, p38MAPK inhibitor SB203580, or the ERK inhibitor PD0325901 alone had minimal influence on RVFV titers. The combination of PF-4708671 and BI-D1870 treatment resulted in robust inhibition of RVFV replication. Likewise, a synergistic inhibition of RVFV replication was observed with p38MAPK inhibitor SB203580 or the ERK inhibitor PD0325901 combined with rapamycin treatment. These findings serve as a proof of concept regarding combination kinase inhibitor treatment for RVFV infection.


Asunto(s)
Antivirales/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Virus de la Fiebre del Valle del Rift/fisiología , Replicación Viral/efectos de los fármacos , Animales , Línea Celular , Ratones , Fosforilación , Biosíntesis de Proteínas/efectos de los fármacos , Procesamiento Proteico-Postraduccional , Proteínas Ribosómicas/metabolismo
11.
Sci Rep ; 8(1): 1925, 2018 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-29386590

RESUMEN

Rift Valley fever virus (RVFV) is a mosquito-borne hemorrhagic fever virus affecting both humans and animals with severe morbidity and mortality and is classified as a potential bioterror agent due to the possible aerosol transmission. At present there is no human vaccine or antiviral therapy available. Thus, there is a great need to develop new antivirals for treatment of RVFV infections. Benzavir-2 was previously identified as potent inhibitor of human adenovirus, herpes simplex virus type 1, and type 2. Here we assess the anti-RVFV activity of benzavir-2 together with four structural analogs and determine pre-clinical pharmacokinetic parameters of benzavir-2. In vitro, benzavir-2 efficiently inhibited RVFV infection, viral RNA production and production of progeny viruses. In vitro, benzavir-2 displayed satisfactory solubility, good permeability and metabolic stability. In mice, benzavir-2 displayed oral bioavailability with adequate maximum serum concentration. Oral administration of benzavir-2 formulated in peanut butter pellets gave high systemic exposure without any observed toxicity in mice. To summarize, our data demonstrated potent anti-RVFV activity of benzavir-2 in vitro together with a promising pre-clinical pharmacokinetic profile. This data support further exploration of the antiviral activity of benzavir-2 in in vivo efficacy models that may lead to further drug development for human use.


Asunto(s)
Antivirales/farmacología , Antivirales/farmacocinética , Benzoatos/farmacología , Benzoatos/farmacocinética , Virus de la Fiebre del Valle del Rift/fisiología , Células A549 , Administración Oral , Animales , Antivirales/administración & dosificación , Antivirales/química , Benzoatos/administración & dosificación , Benzoatos/química , Disponibilidad Biológica , Femenino , Humanos , Ratones Endogámicos BALB C , ARN Viral/genética , Fiebre del Valle del Rift/tratamiento farmacológico , Fiebre del Valle del Rift/prevención & control , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/efectos de los fármacos
12.
J Virol ; 91(21)2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28794043

RESUMEN

There is an urgent need for therapeutic development to combat infections caused by Rift Valley fever virus (RVFV), which causes devastating disease in both humans and animals. In an effort to repurpose drugs for RVFV treatment, our previous studies screened a library of FDA-approved drugs. The most promising candidate identified was the hepatocellular and renal cell carcinoma drug sorafenib. Mechanism-of-action studies indicated that sorafenib targeted a late stage in virus infection and caused a buildup of virions within cells. In addition, small interfering RNA (siRNA) knockdown studies suggested that nonclassical targets of sorafenib are important for the propagation of RVFV. Here we extend our previous findings to identify the mechanism by which sorafenib inhibits the release of RVFV virions from the cell. Confocal microscopy imaging revealed that glycoprotein Gn colocalizes and accumulates within the endoplasmic reticulum (ER) and the transport of Gn from the Golgi complex to the host cell membrane is reduced. Transmission electron microscopy demonstrated that sorafenib caused virions to be present inside large vacuoles inside the cells. p97/valosin-containing protein (VCP), which is involved in membrane remodeling in the secretory pathway and a known target of sorafenib, was found to be important for RVFV egress. Knockdown of VCP resulted in decreased RVFV replication, reduced Gn Golgi complex localization, and increased Gn ER accumulation. The intracellular accumulation of RVFV virions was also observed in cells transfected with siRNA targeting VCP. Collectively, these data indicate that sorafenib causes a disruption in viral egress by targeting VCP and the secretory pathway, resulting in a buildup of virions within dilated ER vesicles.IMPORTANCE In humans, symptoms of RVFV infection mainly include a self-limiting febrile illness. However, in some cases, infected individuals can also experience hemorrhagic fever, neurological disorders, liver failure, and blindness, which could collectively be lethal. The ability of RVFV to expand geographically outside sub-Saharan Africa is of concern, particularly to the Americas, where native mosquito species are capable of virus transmission. Currently, there are no FDA-approved therapeutics to treat RVFV infection, and thus, there is an urgent need to understand the mechanisms by which the virus hijacks the host cell machinery to replicate. The significance of our research is in identifying the cellular target of sorafenib that inhibits RVFV propagation, so that this information can be used as a tool for the further development of therapeutics used to treat RVFV infection.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Fiebre del Valle del Rift/tratamiento farmacológico , Virus de la Fiebre del Valle del Rift/fisiología , Vías Secretoras/efectos de los fármacos , Liberación del Virus/efectos de los fármacos , Adenosina Trifosfatasas/genética , Animales , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/virología , Proteínas de Ciclo Celular/genética , Chlorocebus aethiops , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/virología , Niacinamida/farmacología , Fiebre del Valle del Rift/metabolismo , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Sorafenib , Células Tumorales Cultivadas , Proteína que Contiene Valosina , Células Vero , Virión/efectos de los fármacos , Replicación Viral/efectos de los fármacos
13.
J Virol ; 91(13)2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28381571

RESUMEN

A dynamic actin cytoskeleton is necessary for viral entry, intracellular migration, and virion release. For HIV-1 infection, during entry, the virus triggers early actin activity by hijacking chemokine coreceptor signaling, which activates a host dependency factor, cofilin, and its kinase, the LIM domain kinase (LIMK). Although knockdown of human LIM domain kinase 1 (LIMK1) with short hairpin RNA (shRNA) inhibits HIV infection, no specific small-molecule inhibitor of LIMK has been available. Here, we describe the design and discovery of novel classes of small-molecule inhibitors of LIMK for inhibiting HIV infection. We identified R10015 as a lead compound that blocks LIMK activity by binding to the ATP-binding pocket. R10015 specifically blocks viral DNA synthesis, nuclear migration, and virion release. In addition, R10015 inhibits multiple viruses, including Zaire ebolavirus (EBOV), Rift Valley fever virus (RVFV), Venezuelan equine encephalitis virus (VEEV), and herpes simplex virus 1 (HSV-1), suggesting that LIMK inhibitors could be developed as a new class of broad-spectrum antiviral drugs.IMPORTANCE The actin cytoskeleton is a structure that gives the cell shape and the ability to migrate. Viruses frequently rely on actin dynamics for entry and intracellular migration. In cells, actin dynamics are regulated by kinases, such as the LIM domain kinase (LIMK), which regulates actin activity through phosphorylation of cofilin, an actin-depolymerizing factor. Recent studies have found that LIMK/cofilin are targeted by viruses such as HIV-1 for propelling viral intracellular migration. Although inhibiting LIMK1 expression blocks HIV-1 infection, no highly specific LIMK inhibitor is available. This study describes the design, medicinal synthesis, and discovery of small-molecule LIMK inhibitors for blocking HIV-1 and several other viruses and emphasizes the feasibility of developing LIMK inhibitors as broad-spectrum antiviral drugs.


Asunto(s)
Antivirales/farmacología , Inhibidores Enzimáticos/farmacología , VIH-1/efectos de los fármacos , Quinasas Lim/antagonistas & inhibidores , Liberación del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Antivirales/síntesis química , Antivirales/aislamiento & purificación , Células Cultivadas , Ebolavirus/efectos de los fármacos , Virus de la Encefalitis Equina Venezolana/efectos de los fármacos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/aislamiento & purificación , VIH-1/fisiología , Herpesvirus Humano 1/efectos de los fármacos , Humanos , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Virus de la Fiebre del Valle del Rift/efectos de los fármacos
14.
Antiviral Res ; 143: 162-175, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28442428

RESUMEN

Despite over 60 years of research on antiviral drugs, very few are FDA approved to treat acute viral infections. Rift Valley fever virus (RVFV), an arthropod borne virus that causes hemorrhagic fever in severe cases, currently lacks effective treatments. Existing as obligate intracellular parasites, viruses have evolved to manipulate host cell signaling pathways to meet their replication needs. Specifically, translation modulation is often necessary for viruses to establish infection in their host. Here we demonstrated phosphorylation of p70 S6 kinase, S6 ribosomal protein, and eIF4G following RVFV infection in vitro through western blot analysis and in a mouse model of infection through reverse phase protein microarrays (RPPA). Inhibition of p70 S6 kinase through rapamycin treatment reduced viral titers in vitro and increased survival and mitigated clinical disease in RVFV challenged mice. Additionally, the phosphorylation of p70 S6 kinase was decreased following rapamycin treatment in vivo. Collectively these data demonstrate modulating p70 S6 kinase can be an effective antiviral strategy.


Asunto(s)
Proteínas Quinasas S6 Ribosómicas 70-kDa/efectos de los fármacos , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sirolimus/antagonistas & inhibidores , Animales , Antivirales/farmacología , Apoptosis/efectos de los fármacos , Línea Celular , Chlorocebus aethiops , Replicación del ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Factor 4G Eucariótico de Iniciación/metabolismo , Femenino , Inmunohistoquímica , Hígado/patología , Ratones , Ratones Endogámicos BALB C , Fosforilación/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Fiebre del Valle del Rift/tratamiento farmacológico , Fiebre del Valle del Rift/patología , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/genética , Virus de la Fiebre del Valle del Rift/crecimiento & desarrollo , Virus de la Fiebre del Valle del Rift/patogenicidad , Sirolimus/metabolismo , Sirolimus/uso terapéutico , Análisis de Supervivencia , Células Vero , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
15.
Virology ; 496: 175-185, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27318793

RESUMEN

Rift Valley fever (RVF) is a zoonotic disease that can cause severe illness in humans and livestock, triggering spontaneous abortion in almost 100% of pregnant ruminants. In this study, we demonstrate that signal transducer and activator of transcription 3 (STAT3) is phosphorylated on its conserved tyrosine residue (Y705) following RVFV infection. This phosphorylation was dependent on a major virulence factor, the viral nonstructural protein NSs. Loss of STAT3 had little effect on viral replication, but rather resulted in cells being more susceptible to RVFV-induced cell death. Phosphorylated STAT3 translocated to the nucleus, coinciding with inhibition of fos, jun, and nr4a2 gene expression, and the presence of STAT3 and NSs at the nr4a2 promoter. NSs was found predominantly in the cytoplasm of STAT3 null cells, indicating that STAT3 influences NSs nuclear localization. Collectively, these data demonstrate that STAT3 functions in a pro-survival capacity through modulation of NSs localization.


Asunto(s)
Fiebre del Valle del Rift/metabolismo , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/fisiología , Factor de Transcripción STAT3/metabolismo , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , Chlorocebus aethiops , Humanos , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , Transporte de Proteínas , Fiebre del Valle del Rift/genética , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Tirosina/metabolismo , Células Vero , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
16.
Nanomedicine ; 12(5): 1185-92, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26970026

RESUMEN

In this work we have tested the potential antiviral activity of silver nanoparticles formulated as Argovit™ against Rift Valley fever virus (RVFV). The antiviral activity of Argovit was tested on Vero cell cultures and in type-I interferon receptor deficient mice (IFNAR (-/-) mice) by two different approaches: (i) different dilutions of Argovit were added to previously infected cells or administrated to animals infected with a lethal dose of virus; (ii) virus was pre-incubated with different dilutions of Argovit before inoculation in mice or cells. Though the ability of silver nanoparticles to control an ongoing RVFV infection in the conditions tested was limited, the incubation of virus with Argovit before the infection led to a reduction of the infectivity titers both in vitro and in vivo. These results reveal the potential application of silver nanoparticles to control the infectivity of RVFV, which is an important zoonotic pathogen.


Asunto(s)
Antivirales/farmacología , Nanopartículas/uso terapéutico , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Plata/uso terapéutico , Animales , Ratones , Fiebre del Valle del Rift/prevención & control , Virus de la Fiebre del Valle del Rift/patogenicidad
17.
J Biomol Screen ; 21(4): 354-62, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26762502

RESUMEN

Rift Valley fever virus (RVFV) is an emerging virus that causes serious illness in humans and livestock. There are no approved vaccines or treatments for humans. The purpose of the study was to identify inhibitory compounds of RVFV infection without any preconceived idea of the mechanism of action. A whole-cell-based high-throughput drug screening assay was developed to screen 28,437 small chemical compounds targeting RVFV infection. To accomplish both speed and robustness, a replication-competent NSs-deleted RVFV expressing a fluorescent reporter gene was developed. Inhibition of fluorescence intensity was quantified by spectrophotometry and related to virus infection in human lung epithelial cells (A549). Cell toxicity was assessed by the Resazurin cell viability assay. After primary screening, 641 compounds were identified that inhibited RVFV infection by ≥80%, with ≥50% cell viability at 50 µM concentration. These compounds were subjected to a second screening regarding dose-response profiles, and 63 compounds with ≥60% inhibition of RVFV infection at 3.12 µM compound concentration and ≥50% cell viability at 25 µM were considered hits. Of these, six compounds with high inhibitory activity were identified. In conclusion, the high-throughput assay could efficiently and safely identify several promising compounds that inhibited RVFV infection.


Asunto(s)
Antivirales/farmacología , Benzamidas/farmacología , Ensayos Analíticos de Alto Rendimiento , Hidrazonas/farmacología , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Células A549 , Antivirales/química , Benzamidas/química , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Expresión Génica , Genes Reporteros , Humanos , Hidrazonas/química , Proteínas Luminiscentes/antagonistas & inhibidores , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Virus Reordenados , Virus de la Fiebre del Valle del Rift/crecimiento & desarrollo , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad , Replicación Viral/efectos de los fármacos , Proteína Fluorescente Roja
18.
Antiviral Res ; 127: 79-89, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26801627

RESUMEN

Rift Valley fever virus (RVFV), genus Phlebovirus family Bunyaviridae, is an arthropod-borne virus endemic throughout sub-Saharan Africa. Recent outbreaks have resulted in cyclic epidemics with an increasing geographic footprint, devastating both livestock and human populations. Despite being recognized as an emerging threat, relatively little is known about the virulence mechanisms and host interactions of RVFV. To date there are no FDA approved therapeutics or vaccines for RVF and there is an urgent need for their development. The Ser/Thr protein phosphatase 1 (PP1) has previously been shown to play a significant role in the replication of several viruses. Here we demonstrate for the first time that PP1 plays a prominent role in RVFV replication early on during the viral life cycle. Both siRNA knockdown of PP1α and a novel PP1-targeting small molecule compound 1E7-03, resulted in decreased viral titers across several cell lines. Deregulation of PP1 was found to inhibit viral RNA production, potentially through the disruption of viral RNA transcript/protein interactions, and indicates a potential link between PP1α and the viral L polymerase and nucleoprotein. These results indicate that PP1 activity is important for RVFV replication early on during the viral life cycle and may prove an attractive therapeutic target.


Asunto(s)
Antivirales/farmacología , Replicación del ADN/fisiología , Indoles/farmacología , Proteína Fosfatasa 1/metabolismo , Virus de la Fiebre del Valle del Rift/enzimología , Virus de la Fiebre del Valle del Rift/fisiología , Urea/análogos & derivados , Replicación Viral/fisiología , Animales , Antivirales/química , Antivirales/uso terapéutico , Línea Celular , Replicación del ADN/efectos de los fármacos , Genoma Viral/efectos de los fármacos , Interacciones Huésped-Patógeno , Humanos , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Proteína Fosfatasa 1/antagonistas & inhibidores , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , ARN Viral/biosíntesis , ARN Viral/efectos de los fármacos , Fiebre del Valle del Rift/tratamiento farmacológico , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Virus de la Fiebre del Valle del Rift/genética , Urea/farmacología , Células Vero , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/metabolismo , Virulencia , Replicación Viral/efectos de los fármacos
19.
J Egypt Soc Parasitol ; 45(1): 193-8, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26012234

RESUMEN

This study monitored the antiviral potential of bee venom and four wax extracts, ethanol white and black beeswax (EWW/EBW) and acetone white and black beeswax (AWW/ABW) extracts. Two different virus models namely Adeno-7 as DNA model and RVFV as RNA virus models. End point calculation assay was used to calculate virus depletion titer. The depletion of viral infectivity titer of ABW to Adeno-7 virus showed strong antiviral activity recorded a depletion of viral infectivity titer (1.66 log (10)/ ml) that gave equal action with bee venom and more than interferon IFN (1 log (10)/ ml). On the other hand, antiviral activity of EBW showed a moderate potential, while AWW showed no antiviral activity. Finally EWW showed synergetic activity against Adeno-7 virus activity. Thus, activity of wax extracts to RVFV was arranged in order of IFN bee venom > AWW & EBW > EWW and ABW recorded 3.34, 0.65, 0.5, 0.34 respectively. It is the first time to study the beeswax effect against DNA and RNA virus' models; acetone black beeswax recorded a depletion titer 1.66 log (10)/ml.


Asunto(s)
Antivirales/farmacología , Venenos de Abeja/farmacología , Virus ADN/efectos de los fármacos , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Ceras/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Células Vero
20.
Antiviral Res ; 120: 48-56, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26001632

RESUMEN

Rift Valley Fever Virus (RVFV) belongs to the family Bunyaviridae and is a known cause of epizootics and epidemics in Africa and the Middle East. With no FDA approved therapeutics available to treat RVFV infection, understanding the interactions between the virus and the infected host is crucial to developing novel therapeutic strategies. Here, we investigated the requirement of the ubiquitin-proteasome system (UPS) for the establishment of a productive RVFV infection. It was previously shown that the UPS plays a central role in RVFV multiplication involving degradation of PKR and p62 subunit of TFIIH. Using the FDA-approved proteasome inhibitor Bortezomib, we observed robust inhibition of intracellular and extracellular viral loads. Bortezomib treatment did not affect the nuclear/cytoplasmic distribution of the non-structural S-segment protein (NSs); however, the ability of NSs to form nuclear filaments was abolished as a result of Bortezomib treatment. In silico ubiquitination prediction analysis predicted that known NSs interactors (SAP30, YY1, and mSin3A) have multiple putative ubiquitination sites, while NSs itself was not predicted to be ubiquitinated. Immunoprecipitation studies indicated a decrease in interaction between SAP30 - NSs, and mSin3A - NSs in the context of Bortezomib treatment. This decrease in association between SAP30 - NSs also correlated with a decrease in the ubiquitination status of SAP30 with Bortezomib treatment. Bortezomib treatment, however, resulted in increased ubiquitination of mSin3A, suggesting that Bortezomib dynamically affects the ubiquitination status of host proteins that interact with NSs. Finally, we observed that expression of interferon beta (IFN-ß) was increased in Bortezomib treated cells which indicated that the cellular antiviral mechanism was revived as a result of treatment and may contribute to control of viral multiplication.


Asunto(s)
Antivirales/farmacología , Bortezomib/farmacología , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Virus de la Fiebre del Valle del Rift/fisiología , Replicación Viral/efectos de los fármacos , Células Cultivadas , Células Epiteliales/virología , Humanos , Carga Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...