Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Viruses ; 14(2)2022 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-35216015

RESUMO

Mayaro virus (MAYV) manipulates cell machinery to successfully replicate. Thus, identifying host proteins implicated in MAYV replication represents an opportunity to discover potential antiviral targets. PIM kinases are enzymes that regulate essential cell functions and also appear to be critical factors in the replication of certain viruses. In this study we explored the consequences of PIM kinase inhibition in the replication of MAYV and other arboviruses. Cytopathic effects or viral titers in samples from MAYV-, Chikungunya-, Una- or Zika-infected cells treated with PIM kinase inhibitors were evaluated using an inverted microscope or plaque-forming assays. The expression of viral proteins E1 and nsP1 in MAYV-infected cells was assessed using an immunofluorescence confocal microscope or Western blot. Our results revealed that PIM kinase inhibition partially prevented MAYV-induced cell damage and also promoted a decrease in viral titers for MAYV, UNAV and ZIKV. The inhibitory effect of PIM kinase blocking was observed for each of the MAYV strains tested and also occurred as late as 8 h post infection (hpi). Finally, PIM kinase inhibition suppressed the expression of MAYV E1 and nsP1 proteins. Taken together, these findings suggest that PIM kinases could represent an antiviral target for MAYV and other arboviruses.


Assuntos
Alphavirus/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-pim-1/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos , Animais , Antivirais/farmacologia , Linhagem Celular , Vírus Chikungunya/efeitos dos fármacos , Humanos , Zika virus/efeitos dos fármacos
2.
J Virol ; 96(2): e0177421, 2022 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-34757841

RESUMO

Alphaviruses and flaviviruses have class II fusion glycoproteins that are essential for virion assembly and infectivity. Importantly, the tip of domain II is structurally conserved between the alphavirus and flavivirus fusion proteins, yet whether these structural similarities between virus families translate to functional similarities is unclear. Using in vivo evolution of Zika virus (ZIKV), we identified several novel emerging variants, including an envelope glycoprotein variant in ß-strand c (V114M) of domain II. We have previously shown that the analogous ß-strand c and the ij loop, located in the tip of domain II of the alphavirus E1 glycoprotein, are important for infectivity. This led us to hypothesize that flavivirus E ß-strand c also contributes to flavivirus infection. We generated this ZIKV glycoprotein variant and found that while it had little impact on infection in mosquitoes, it reduced replication in human cells and mice and increased virus sensitivity to ammonium chloride, as seen for alphaviruses. In light of these results and given our alphavirus ij loop studies, we mutated a conserved alanine at the tip of the flavivirus ij loop to valine to test its effect on ZIKV infectivity. Interestingly, this mutation inhibited infectious virion production of ZIKV and yellow fever virus, but not West Nile virus. Together, these studies show that shared domains of the alphavirus and flavivirus class II fusion glycoproteins harbor structurally analogous residues that are functionally important and contribute to virus infection in vivo.IMPORTANCE Arboviruses are a significant global public health threat, yet there are no antivirals targeting these viruses. This problem is in part due to our lack of knowledge of the molecular mechanisms involved in the arbovirus life cycle. In particular, virus entry and assembly are essential processes in the virus life cycle and steps that can be targeted for the development of antiviral therapies. Therefore, understanding common, fundamental mechanisms used by different arboviruses for entry and assembly is essential. In this study, we show that flavivirus and alphavirus residues located in structurally conserved and analogous regions of the class II fusion proteins contribute to common mechanisms of entry, dissemination, and infectious-virion production. These studies highlight how class II fusion proteins function and provide novel targets for development of antivirals.


Assuntos
Alphavirus/fisiologia , Flavivirus/fisiologia , Proteínas Virais de Fusão/metabolismo , Vírion/metabolismo , Replicação Viral , Células A549 , Alphavirus/efeitos dos fármacos , Cloreto de Amônio/farmacologia , Animais , Culicidae/virologia , Flavivirus/efeitos dos fármacos , Humanos , Interferon Tipo I/deficiência , Camundongos , Camundongos Mutantes , Mutação , Domínios Proteicos , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Vírion/genética , Montagem de Vírus/genética , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/genética , Zika virus/efeitos dos fármacos , Zika virus/fisiologia , Infecção por Zika virus/virologia
3.
Viruses ; 11(4)2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-31018496

RESUMO

Mayaro (MAYV) and Una (UNAV) are emerging arboviruses belonging to the Alphavirus genus of the Togaviridae family. These viruses can produce febrile disease with symptoms such as fever, headache, myalgia, skin rash and incapacitating poly-arthralgia. Serological studies indicate that both viruses are circulating in different countries in Latin America. Viruses need the host cell machinery and resources to replicate effectively. One strategy to find new antivirals consists of identifying key cellular pathways or factors that are essential for virus replication. In this study, we analyzed the role of the ubiquitin-proteasome system (UPS) in MAYV and UNAV replication. Vero-E6 or HeLa cells were treated with the proteasome inhibitors MG132 or Lactacystin, and viral progeny production was quantified using a plaque assay method. In addition, the synthesis of viral proteins was analyzed by Western blot and confocal microscopy. Our results indicate that treatment with proteasome inhibitors decreases MAYV and UNAV protein synthesis, and also causes a significant dose-dependent decrease in MAYV and UNAV replication. Proteasome activity seems to be important at the early stages of MAYV replication. These findings suggest that the ubiquitin-proteasome system is a possible pharmacological target to inhibit these neglected alphaviruses.


Assuntos
Alphavirus/efeitos dos fármacos , Antivirais/farmacologia , Complexo de Endopeptidases do Proteassoma/fisiologia , Replicação Viral , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Alphavirus/fisiologia , Animais , Chlorocebus aethiops , Inibidores de Cisteína Proteinase/farmacologia , Citoplasma/efeitos dos fármacos , Citoplasma/virologia , Células HeLa , Humanos , Leupeptinas/farmacologia , Inibidores de Proteassoma/farmacologia , Células Vero
4.
J Virol ; 93(7)2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30674629

RESUMO

Tetherin is an interferon-inducible, antiviral host factor that broadly restricts enveloped virus release by tethering budded viral particles to the plasma membrane. In response, many viruses have evolved tetherin antagonists. The human tetherin gene can express two isoforms, long and short, due to alternative translation initiation sites in the N-terminal cytoplasmic tail. The long isoform (L-tetherin) contains 12 extra amino acids in its N terminus, including a dual tyrosine motif (YDYCRV) that is an internalization signal for clathrin-mediated endocytosis and a determinant of NF-κB activation. Tetherin restricts alphaviruses, which are highly organized enveloped RNA viruses that bud from the plasma membrane. L-tetherin is more efficient than S-tetherin in inhibiting alphavirus release in 293 cells. Here, we demonstrated that alphaviruses do not encode an antagonist for either of the tetherin isoforms. Instead, the isoform specificity reflected a requirement for tetherin endocytosis. The YXY motif in L-tetherin was necessary for alphavirus restriction in 293 cells but was not required for rhabdovirus restriction. L-tetherin's inhibition of alphavirus release correlated with its internalization but did not involve NF-κB activation. In contrast, in U-2 OS cells, the YXY motif and the L-tetherin N-terminal domain were not required for either robust tetherin internalization or alphavirus inhibition. Tetherin forms that were negative for restriction accumulated at the surface of infected cells, while the levels of tetherin forms that restrict were decreased. Together, our results suggest that tetherin-mediated virus internalization plays an important role in the restriction of alphavirus release and that cell-type-specific cofactors may promote tetherin endocytosis.IMPORTANCE The mechanisms of tetherin's antiviral activities and viral tetherin antagonism have been studied in detail for a number of different viruses. Although viral countermeasures against tetherin can differ significantly, overall, tetherin's antiviral activity correlates with physical tethering of virus particles to prevent their release. While tetherin can mediate virus endocytic uptake and clearance, this has not been observed to be required for restriction. Here we show that efficient tetherin inhibition of alphavirus release requires efficient tetherin endocytosis. Our data suggest that this endocytic uptake can be mediated by tetherin itself or by a tetherin cofactor that promotes uptake of an endocytosis-deficient variant of tetherin.


Assuntos
Alphavirus/efeitos dos fármacos , Antígeno 2 do Estroma da Médula Óssea/farmacologia , Liberação de Vírus/efeitos dos fármacos , Infecções por Alphavirus/tratamento farmacológico , Infecções por Alphavirus/metabolismo , Infecções por Alphavirus/virologia , Animais , Linhagem Celular , Cricetinae , Endocitose/efeitos dos fármacos , Células HEK293 , Humanos , NF-kappa B/metabolismo , Isoformas de Proteínas/metabolismo , Vírion/efeitos dos fármacos
5.
Rev Soc Bras Med Trop ; 51(5): 584-590, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30304262

RESUMO

INTRODUCTION: The Mayaro virus (MAYV), which is an arbovirus closely related to the Chikungunya virus, causes a dengue-like acute illness that is endemic to Central and South America. We investigated the anti-MAYV activity of prostaglandin A1 (PGA1), a hormone which exhibits antiviral activity against both ribonucleic acid (RNA) and deoxyribonucleic acid (DNA) viruses. Further, we examined the effects of inducting the stress protein HSP70 following PGA1 treatment. METHODS: Hep-2 cells infected with MAYV were treated with PGA1 (0.1-6µg/ml) 12h before infection and for different periods post-infection. Inhibition of viral replication inhibition was analyzed via viral titer determination, whereas the effect of PGA1 on viral morphogenesis was examined via transmission electron microscopy (TEM). Autoradiography (with 35S methionine labeling) and western blotting were used to assess the effect of PGA1 treatment on viral and cellular protein synthesis, and on HSP70 induction, respectively. RESULTS: PGA1 strongly reduced viral replication in Hep-2 cells, particularly when added during the early stages of viral replication. Although PGA1 treatment inhibited viral replication by 95% at 24 hours post-infection (hpi), viral structural protein synthesis was inhibited only by 15%. TEM analysis suggested that PGA1 inhibited replication before viral morphogenesis. Western blot and densitometry analyses showed that PGA1 treatment increased HSP70 protein levels, although this was not detectable via autoradiography. CONCLUSIONS: PGA1 inhibits MAYV replication in Hep-2 cells at early stages of viral replication, prior to production of viral structural proteins, possibly via HSP70 induction.


Assuntos
Alphavirus/efeitos dos fármacos , Células Epiteliais/virologia , Proteínas de Choque Térmico HSP70/farmacologia , Prostaglandinas A/farmacologia , Replicação Viral/efeitos dos fármacos , Alphavirus/ultraestrutura , Animais , Antivirais/farmacologia , Western Blotting , Bovinos , Linhagem Celular , Eletroforese em Gel de Poliacrilamida , Células Epiteliais/ultraestrutura , Humanos , Microscopia Eletrônica de Transmissão
6.
Rev. Soc. Bras. Med. Trop ; 51(5): 584-590, Sept.-Oct. 2018. tab, graf
Artigo em Inglês | LILACS | ID: biblio-957459

RESUMO

Abstract INTRODUCTION: The Mayaro virus (MAYV), which is an arbovirus closely related to the Chikungunya virus, causes a dengue-like acute illness that is endemic to Central and South America. We investigated the anti-MAYV activity of prostaglandin A1 (PGA1), a hormone which exhibits antiviral activity against both ribonucleic acid (RNA) and deoxyribonucleic acid (DNA) viruses. Further, we examined the effects of inducting the stress protein HSP70 following PGA1 treatment. METHODS: Hep-2 cells infected with MAYV were treated with PGA1 (0.1-6μg/ml) 12h before infection and for different periods post-infection. Inhibition of viral replication inhibition was analyzed via viral titer determination, whereas the effect of PGA1 on viral morphogenesis was examined via transmission electron microscopy (TEM). Autoradiography (with 35S methionine labeling) and western blotting were used to assess the effect of PGA1 treatment on viral and cellular protein synthesis, and on HSP70 induction, respectively. RESULTS: PGA1 strongly reduced viral replication in Hep-2 cells, particularly when added during the early stages of viral replication. Although PGA1 treatment inhibited viral replication by 95% at 24 hours post-infection (hpi), viral structural protein synthesis was inhibited only by 15%. TEM analysis suggested that PGA1 inhibited replication before viral morphogenesis. Western blot and densitometry analyses showed that PGA1 treatment increased HSP70 protein levels, although this was not detectable via autoradiography. CONCLUSIONS: PGA1 inhibits MAYV replication in Hep-2 cells at early stages of viral replication, prior to production of viral structural proteins, possibly via HSP70 induction.


Assuntos
Humanos , Animais , Bovinos , Prostaglandinas A/farmacologia , Replicação Viral/efeitos dos fármacos , Alphavirus/efeitos dos fármacos , Proteínas de Choque Térmico HSP70/farmacologia , Células Epiteliais/virologia , Antivirais/farmacologia , Linhagem Celular , Western Blotting , Alphavirus/ultraestrutura , Microscopia Eletrônica de Transmissão , Eletroforese em Gel de Poliacrilamida , Células Epiteliais/ultraestrutura
7.
Antiviral Res ; 158: 8-12, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30076863

RESUMO

Mayaro virus (MAYV) is a neglected arbovirus belonging to the family Togaviridae. Its infection leads to Mayaro fever, with clinical manifestations such as fever, myalgia, headache, rash, arthralgia, vomiting, and diarrhea. The most prominent complaint from infected person is the long-lasting arthritis/arthralgia. The treatment for Mayaro fever is mainly symptom-based and there are no vaccines or antiviral drugs currently available, thus, natural products with anti-MAYV activity may provide a potential alternative. Recent evidences suggest that oxidative stress plays an important role in MAYV infection and compounds capable of modulating oxidative stress could represent a novel therapeutic approach in modulating MAYV-associated oxidative cellular damage. Silymarin is a complex extracted of Silybum marianum, or milk thistle, and its major active compound is silybin, which has a remarkable biological effect. Its antioxidant and antiviral effects, including its antiviral activity against the Chikungunya virus (CHIKV), prompted us to think whether silymarin could also reduce the replication of the MAYV and restore the pro-oxidant/antioxidant balance in the context of MAYV infection, leading to reduced cellular oxidative stress. We assessed the antiviral activity and protective effect of silymarin against oxidative stress in MAYV-infected HepG2 cells. Cytopathic effect inhibition, viral replication, and plaque reduction assays were used to determine the anti-MAYV activity of silymarin. Additionally, we determined whether silymarin could reduce MAYV-induced oxidative cell damage. Briefly, silymarin exhibited potent antiviral activity against MAYV and reduced MAYV-induced ROS formation and levels of malondialdehyde (MDA) and carbonyl protein, which are biomarkers of oxidative stress. In conclusion, the ability of silymarin to inhibit MAYV replication and attenuate MAYV-induce oxidative stress warrants further investigation of this compound as a novel therapeutic approach to Mayaro fever disease.


Assuntos
Alphavirus/efeitos dos fármacos , Antivirais/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Silimarina/farmacologia , Infecções por Alphavirus/tratamento farmacológico , Antioxidantes/farmacologia , Vírus Chikungunya/efeitos dos fármacos , Células Hep G2 , Humanos , Silybum marianum/química , Espécies Reativas de Oxigênio , Silibina/farmacologia , Replicação Viral/efeitos dos fármacos
8.
Antiviral Res ; 157: 57-67, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29981794

RESUMO

The New World alphaviruses -Venezuelan, eastern, and western equine encephalitis viruses (VEEV, EEEV, and WEEV respectively) - cause a febrile disease that is often lethal in equines and children and leads to long-term neurological sequelae in survivors. Endemic to the Americas, epizootic outbreaks of the three viruses occur sporadically in the continental United States. All three viruses aerosolize readily, replicate to high titers in cell culture, and have low infectious doses. Additionally, there are no FDA-approved vaccines or therapeutics for human use. To address the therapeutic gap, a high throughput assay utilizing a luciferase reporter virus, TC83-luc, was performed to screen a library of commercially available, FDA-approved drugs for antiviral activity. From a group of twenty compounds found to significantly decrease luminescence, the carcinoma therapeutic sorafenib inhibited replication of VEEV-TC83 and TrD in vitro. Additionally, sorafenib inhibited replication of EEEV and two Old World alphaviruses, Sindbis virus and chikungunya virus, at 8 and 16 h post-infection. Sorafenib caused no toxicity in Vero cells, and coupled with a low EC50 value, yielded a selectivity index of >19. Mechanism of actions studies suggest that sorafenib inhibited viral translation through dephosphorylation of several key proteins, including eIF4E and p70S6K, leading to a reduction in viral protein production and overall viral replication.


Assuntos
Alphavirus/efeitos dos fármacos , Antineoplásicos/farmacologia , Antivirais/farmacologia , Reposicionamento de Medicamentos , Sorafenibe/farmacologia , Replicação Viral/efeitos dos fármacos , Alphavirus/crescimento & desenvolvimento , Animais , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos/métodos , Genes Reporter , Ensaios de Triagem em Larga Escala , Luciferases/análise , Luciferases/genética , Medições Luminescentes , Genética Reversa
9.
Nat Microbiol ; 2(10): 1435-1445, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28848230

RESUMO

Mosquito-borne viruses can cause severe inflammatory diseases and there are limited therapeutic solutions targeted specifically at virus-induced inflammation. Chikungunya virus (CHIKV), a re-emerging alphavirus responsible for several outbreaks worldwide in the past decade, causes debilitating joint inflammation and severe pain. Here, we show that CHIKV infection activates the NLRP3 inflammasome in humans and mice. Peripheral blood mononuclear cells isolated from CHIKV-infected patients showed elevated NLRP3, caspase-1 and interleukin-18 messenger RNA expression and, using a mouse model of CHIKV infection, we found that high NLRP3 expression was associated with peak inflammatory symptoms. Inhibition of NLRP3 activation using the small-molecule inhibitor MCC950 resulted in reduced CHIKV-induced inflammation and abrogated osteoclastogenic bone loss and myositis, but did not affect in vivo viral replication. Mice treated with MCC950 displayed lower expression levels of the cytokines interleukin-6, chemokine ligand 2 and tumour necrosis factor in joint tissue. Interestingly, MCC950 treatment abrogated disease signs in mice infected with a related arthritogenic alphavirus, Ross River virus, but not in mice infected with West Nile virus-a flavivirus. Here, using mouse models of alphavirus-induced musculoskeletal disease, we demonstrate that NLRP3 inhibition in vivo can reduce inflammatory pathology and that further development of therapeutic solutions targeting inflammasome function could help treat arboviral diseases.


Assuntos
Alphavirus/imunologia , Febre de Chikungunya/tratamento farmacológico , Vírus Chikungunya/efeitos dos fármacos , Inflamassomos/farmacologia , Inflamação , Proteína 3 que Contém Domínio de Pirina da Família NLR/efeitos dos fármacos , Alphavirus/efeitos dos fármacos , Animais , Caspase 1 , Quimiocinas/metabolismo , Febre de Chikungunya/patologia , Chlorocebus aethiops , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Interleucina-18/metabolismo , Interleucina-6/metabolismo , Leucócitos Mononucleares/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miosite/patologia , RNA Mensageiro/metabolismo , Ross River virus/efeitos dos fármacos , Células Vero , Vírus do Nilo Ocidental/efeitos dos fármacos
10.
Antiviral Res ; 126: 117-24, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26752081

RESUMO

Chikungunya virus (CHIKV) is an arthritogenic arbovirus of the Alphavirus genus, which has infected millions of people after its re-emergence in the last decade. In this study, a BHK cell line containing a stable CHIKV replicon with a luciferase reporter was used in a high-throughput platform to screen approximately 3000 compounds. Following initial validation, 25 compounds were chosen as primary hits for secondary validation with wild type and reporter CHIKV infection, which identified three promising compounds. Abamectin (EC50 = 1.5 µM) and ivermectin (EC50 = 0.6 µM) are fermentation products generated by a soil dwelling actinomycete, Streptomyces avermitilis, whereas berberine (EC50 = 1.8 µM) is a plant-derived isoquinoline alkaloid. They inhibited CHIKV replication in a dose-dependent manner and had broad antiviral activity against other alphaviruses--Semliki Forest virus and Sindbis virus. Abamectin and ivermectin were also active against yellow fever virus, a flavivirus. These compounds caused reduced synthesis of CHIKV genomic and antigenomic viral RNA as well as downregulation of viral protein expression. Time of addition experiments also suggested that they act on the replication phase of the viral infectious cycle.


Assuntos
Alphavirus/efeitos dos fármacos , Antivirais/farmacologia , Febre de Chikungunya/tratamento farmacológico , Vírus Chikungunya/efeitos dos fármacos , Alphavirus/fisiologia , Animais , Berberina/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Febre de Chikungunya/virologia , Vírus Chikungunya/fisiologia , Cricetinae , DNA Viral/antagonistas & inibidores , DNA Viral/biossíntese , DNA Viral/efeitos dos fármacos , Flavivirus/efeitos dos fármacos , Humanos , Ivermectina/análogos & derivados , Ivermectina/farmacologia , RNA Viral/antagonistas & inibidores , RNA Viral/biossíntese , RNA Viral/efeitos dos fármacos , RNA Viral/genética , Replicon/efeitos dos fármacos , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/biossíntese , Proteínas Virais/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Vírus da Febre Amarela/efeitos dos fármacos
11.
Vet Immunol Immunopathol ; 135(1-2): 93-99, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20004981

RESUMO

Recombinant equine interferon-gamma (reIFN-gamma) was prepared using a baculovirus expression system and its antiviral activity was investigated using several equine viruses. The reIFN-gamma suppressed the replication of all equine viruses used in the present experiment in horse cell cultures, but did not affect the growth of host cells at concentrations of less than 1000 u/ml. A strong antiviral effect was observed, especially against RNA viruses. Equine picornavirus, equine rhinovirus and equine arteritis virus could not be propagated at all in 100 u/ml reIFN-gamma when 100 TCID(50) of infective viruses was inoculated to cultivated horse cells. DNA viruses, equine herpesvirus types 1, 2, 3 and 4 and equine adenovirus, were less sensitive to reIFN-gamma but their growth became less than 1/100 in the cells treated with 100 u/ml reIFN-gamma compared to untreated cells. The antiviral effects were decreased in the cells of heterologous species and more than 1000 u/ml reIFN-gamma was required to induce an antiviral effect.


Assuntos
Adenoviridae/efeitos dos fármacos , Alphavirus/efeitos dos fármacos , Antivirais/farmacologia , Aphthovirus/efeitos dos fármacos , Equartevirus/efeitos dos fármacos , Herpesvirus Equídeo 1/efeitos dos fármacos , Herpesvirus Equídeo 3/efeitos dos fármacos , Herpesvirus Equídeo 4/efeitos dos fármacos , Interferon gama/farmacologia , Picornaviridae/efeitos dos fármacos , Rhadinovirus/efeitos dos fármacos , Animais , Antivirais/efeitos adversos , Células Cultivadas , Relação Dose-Resposta a Droga , Doenças dos Cavalos/tratamento farmacológico , Doenças dos Cavalos/virologia , Cavalos/virologia , Interferon gama/efeitos adversos , Proteínas Recombinantes , Replicação Viral/efeitos dos fármacos
12.
Antiviral Res ; 78(3): 215-22, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18294708

RESUMO

Several members of the widespread alphavirus group are pathogenic, but no therapy is available to treat these RNA virus infections. We report here a quantitative assay to screen for inhibitors of Semliki Forest virus (SFV) replication, and demonstrate the effects of 29 nucleosides on SFV and Sindbis virus replication. The anti-SFV assay developed is based on a SFV strain containing Renilla luciferase inserted after the nsP3 coding region, yielding a marker virus in which the luciferase is cleaved out during polyprotein processing. The reporter-gene assay was miniaturized, automated and validated, resulting in a Z' value of 0.52. [3H]uridine labeling for 1 h at the maximal viral RNA synthesis time point was used as a comparative method. Anti-SFV screening and counter-screening for cell viability led to the discovery of several new SFV inhibitors. 3'-amino-3'-deoxyadenosine was the most potent inhibitor in this set, with an IC50 value of 18 microM in the reporter-gene assay and 2 microM in RNA synthesis rate detection. Besides the 3'-substituted analogues, certain N6-substituted nucleosides had similar IC50 values for both SFV and Sindbis replication, suggesting the applicability of this methodology to alphaviruses in general.


Assuntos
Alphavirus/efeitos dos fármacos , Antivirais , Genes Reporter , Luciferases/metabolismo , Nucleosídeos , Replicação Viral/efeitos dos fármacos , Alphavirus/genética , Alphavirus/fisiologia , Animais , Antivirais/química , Antivirais/farmacologia , Linhagem Celular , Cricetinae , Desoxiadenosinas/química , Desoxiadenosinas/farmacologia , Concentração Inibidora 50 , Luciferases/genética , Testes de Sensibilidade Microbiana/métodos , Nucleosídeos/química , Nucleosídeos/farmacologia , RNA Viral/biossíntese , Vírus da Floresta de Semliki/efeitos dos fármacos , Vírus da Floresta de Semliki/genética , Vírus da Floresta de Semliki/fisiologia , Sindbis virus/efeitos dos fármacos , Sindbis virus/genética , Sindbis virus/fisiologia
13.
Proc Natl Acad Sci U S A ; 104(19): 8083-8, 2007 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-17470783

RESUMO

Plants have evolved multiple mechanisms to selectively suppress pathogens by production of secondary metabolites with antimicrobial activities. Therefore, direct selections for antiviral compounds from plants can be used to identify new agents with potent antiviral activity but not toxic to hosts. Here, we provide evidence that a class of compounds, seco-pregnane steroid glaucogenin C and its monosugar-glycoside cynatratoside A of Strobilanthes cusia and three new pantasugar-glycosides of glaucogenin C of Cynanchum paniculatum, are effective and selective inhibitors to alphavirus-like positive-strand RNA viruses including plant-infecting tobacco mosaic virus (TMV) and animal-infecting Sindbis virus (SINV), eastern equine encephalitis virus, and Getah virus, but not to other RNA or DNA viruses, yet they were not toxic to host cells. In vivo administration of the compounds protected BALB/c mice from lethal SINV infection without adverse effects on the mice. Using TMV and SINV as models, studies on the action mechanism revealed that the compounds predominantly suppress the expression of viral subgenomic RNA(s) without affecting the accumulation of viral genomic RNA. Our work suggested that the viral subgenomic RNA could be a new target for the discovery of antiviral drugs, and that seco-pregnane steroid and its four glycosides found in the two medicinal herbs have the potential for further development as antiviral agents against alphavirus-like positive-strand RNA viruses.


Assuntos
Alphavirus/efeitos dos fármacos , Antivirais/farmacologia , Pregnanos/farmacologia , RNA Viral/antagonistas & inibidores , Alphavirus/genética , Infecções por Alphavirus/tratamento farmacológico , Animais , Linhagem Celular , Cricetinae , Relação Dose-Resposta a Droga , Camundongos , Camundongos Endogâmicos BALB C , Sindbis virus/efeitos dos fármacos , Vírus do Mosaico do Tabaco/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
14.
Braz. j. med. biol. res ; 31(9): 1119-23, sept. 1998. ilus, graf
Artigo em Inglês | LILACS | ID: lil-222958

RESUMO

The effect of prostaglandins (PGA1 and PGB2) on the replication of Mayaro virus was studied in Vero cells. PGA1 and PGB2 antiviral activity was found to be dose-dependent. However, while 10 µg/ml PGB2 inhibited virus yield by 60 percent, at the same dose PGA1 suppressed virus replication by more than 90 percent. SDS-PAGE analysis of [35S]-methionine-labelled proteins showed that PGA1 did not alter cellular protein synthesis. In infected cells, PGA1 slightly inhibited the synthesis of protein C, while drastically inhibiting the synthesis of glycoproteins E1 and E2


Assuntos
Animais , Alphavirus/fisiologia , Prostaglandinas A/farmacologia , Prostaglandinas B/farmacologia , Células Vero/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Infecções por Alphavirus/tratamento farmacológico , Alphavirus/efeitos dos fármacos , Alphavirus/crescimento & desenvolvimento , Glicoproteínas/biossíntese , Metionina/análise , Prostaglandinas A/metabolismo , Prostaglandinas A/uso terapêutico , Prostaglandinas B/metabolismo , Prostaglandinas B/uso terapêutico , Proteína C/biossíntese
15.
Vopr Virusol ; 30(5): 581-4, 1985.
Artigo em Russo | MEDLINE | ID: mdl-3000084

RESUMO

Chick embryo fibroblast cultures of the C/O phenotype (leukemia--free) infected with eastern equine encephalomyelitis (EEE) virus were incubated in the presence of 15 micrograms/ml N-methyl-N-nitro-N-nitrosoguanidine in the culture medium. Seven (5%) temperature-sensitive mutants were isolated from cell homogenates only in those cases where cell cultures before infection had been treated with actinomycin D. The recovered ts mutants are characterized by the marked ts- phenotype and genetic stability. The method of obtaining EEE virus ts mutants under the effect of N-methyl-N-nitro-N-nitrosoguanidine in C/O phenotype (leukemia-free) chick embryo fibroblast cultures treated before virus inoculation with actinomycin D is discussed.


Assuntos
Alphavirus/efeitos dos fármacos , Vírus da Encefalite Equina do Leste/efeitos dos fármacos , Mutação , Temperatura , Animais , Embrião de Galinha , Dactinomicina/farmacologia , Vírus da Encefalite Equina do Leste/isolamento & purificação , Vírus da Encefalite Equina do Leste/fisiologia , Metilnitronitrosoguanidina/farmacologia , Fenótipo , Ensaio de Placa Viral , Cultura de Vírus/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA