Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Nat Commun ; 13(1): 558, 2022 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-35091550

RESUMO

Five New World mammarenaviruses (NWMs) cause life-threatening hemorrhagic fever (HF). Cellular entry by these viruses is mediated by human transferrin receptor 1 (hTfR1). Here, we demonstrate that an antibody (ch128.1/IgG1) which binds the apical domain of hTfR1, potently inhibits infection of attenuated and pathogenic NWMs in vitro. Computational docking of the antibody Fab crystal structure onto the known structure of hTfR1 shows an overlapping receptor-binding region shared by the Fab and the viral envelope glycoprotein GP1 subunit that binds hTfR1, and we demonstrate competitive inhibition of NWM GP1 binding by ch128.1/IgG1 as the principal mechanism of action. Importantly, ch128.1/IgG1 protects hTfR1-expressing transgenic mice against lethal NWM challenge. Additionally, the antibody is well-tolerated and only partially reduces ferritin uptake. Our findings provide the basis for the development of a novel, host receptor-targeted antibody therapeutic broadly applicable to the treatment of HF of NWM etiology.


Assuntos
Antígenos CD/metabolismo , Arenaviridae/metabolismo , Febre Hemorrágica Americana/metabolismo , Receptores da Transferrina/metabolismo , Proteínas do Envelope Viral/metabolismo , Células A549 , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/farmacologia , Antígenos CD/imunologia , Arenaviridae/efeitos dos fármacos , Arenaviridae/fisiologia , Chlorocebus aethiops , Febre Hemorrágica Americana/prevenção & controle , Febre Hemorrágica Americana/virologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Vírus Junin/efeitos dos fármacos , Vírus Junin/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Simulação de Acoplamento Molecular , Ligação Proteica/efeitos dos fármacos , Receptores da Transferrina/antagonistas & inibidores , Receptores da Transferrina/imunologia , Células Vero
2.
Peptides ; 142: 170570, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34000327

RESUMO

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.


Assuntos
Peptídeos/farmacologia , Zoonoses Virais/tratamento farmacológico , Vírus/efeitos dos fármacos , Animais , Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Vírus da Encefalite Equina do Leste/efeitos dos fármacos , Humanos , Vírus Junin/efeitos dos fármacos , Vírus da Febre do Vale do Rift/efeitos dos fármacos , SARS-CoV-2/efeitos dos fármacos
3.
J Gen Virol ; 101(6): 573-586, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32375950

RESUMO

Bone marrow stromal cell antigen-2 (BST-2), also known as tetherin, is an interferon-inducible membrane-associated protein. It effectively targets enveloped viruses at the release step of progeny viruses from host cells, thereby restricting the further spread of viral infection. Junin virus (JUNV) is a member of Arenaviridae, which causes Argentine haemorrhagic fever that is associated with a high rate of mortality. In this study, we examined the effect of human BST-2 on the replication and propagation of JUNV. The production of JUNV Z-mediated virus-like particles (VLPs) was significantly inhibited by over-expression of BST-2. Electron microscopy analysis revealed that BST-2 functions by forming a physical link that directly retains VLPs on the cell surface. Infection using JUNV showed that infectious JUNV production was moderately inhibited by endogenous or exogenous BST-2. We also observed that JUNV infection triggers an intense interferon response, causing an upregulation of BST-2, in infected cells. However, the expression of cell surface BST-2 was reduced upon infection. Furthermore, the expression of JUNV nucleoprotein (NP) partially recovered VLP production from BST-2 restriction, suggesting that the NP functions as an antagonist against antiviral effect of BST-2. We further showed that JUNV NP also rescued the production of Ebola virus VP40-mediated VLP from BST-2 restriction as a broad spectrum BST-2 antagonist. To our knowledge, this is the first report showing that an arenavirus protein counteracts the antiviral function of BST-2.


Assuntos
Antígenos CD/metabolismo , Interações Hospedeiro-Patógeno/fisiologia , Vírus Junin/fisiologia , Nucleoproteínas/metabolismo , Proteínas do Core Viral/metabolismo , Liberação de Vírus/fisiologia , Células A549 , Antivirais/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Proteínas Ligadas por GPI/metabolismo , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Interferons/farmacologia , Vírus Junin/efeitos dos fármacos , Liberação de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
4.
Antiviral Res ; 169: 104558, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31302150

RESUMO

Several mammarenaviruses, chiefly Lassa virus (LASV) in Western Africa and Junín virus (JUNV) in the Argentine Pampas, cause severe disease in humans and pose important public health problems in their endemic regions. Moreover, mounting evidence indicates that the worldwide-distributed mammarenavirus lymphocytic choriomeningitis virus (LCMV) is a neglected human pathogen of clinical significance. The lack of licensed mammarenavirus vaccines and partial efficacy of current anti-mammarenavirus therapy limited to an off-label use of the nucleoside analog ribavirin underscore an unmet need for novel therapeutics to combat human pathogenic mammarenavirus infections. This task can be facilitated by the implementation of "drug repurposing" strategies to reduce the time and resources required to advance identified antiviral drug candidates into the clinic. We screened a drug repurposing library of 11,968 compounds (Repurposing, Focused Rescue and Accelerated Medchem [ReFRAME]) and identified several potent inhibitors of LCMV multiplication that had also strong anti-viral activity against LASV and JUNV. Our findings indicate that enzymes of the rate-limiting steps of pyrimidine and purine biosynthesis, the pro-viral MCL1 apoptosis regulator, BCL2 family member protein and the mitochondrial electron transport complex III, play critical roles in the completion of the mammarenavirus life cycle, suggesting they represent potential druggable targets to counter human pathogenic mammarenavirus infections.


Assuntos
Antivirais/farmacologia , Arenaviridae/efeitos dos fármacos , Bases de Dados de Produtos Farmacêuticos , Avaliação Pré-Clínica de Medicamentos/métodos , Reposicionamento de Medicamentos/métodos , Células A549 , Animais , Apoptose , Arenaviridae/fisiologia , Infecções por Arenaviridae/tratamento farmacológico , Infecções por Arenaviridae/imunologia , Infecções por Arenaviridae/virologia , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Células HEK293 , Humanos , Interferons/genética , Vírus Junin/efeitos dos fármacos , Vírus Lassa/efeitos dos fármacos , Vírus da Coriomeningite Linfocítica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Purinas/biossíntese , Pirimidinas/biossíntese , Células Vero , Replicação Viral/efeitos dos fármacos
5.
Carbohydr Res ; 480: 61-66, 2019 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-31176191

RESUMO

Herein we describe the synthesis of imidazo[2,1-b][1,3,4]thiadiazoles from carbohydrates with D-ribo and D-xylo configuration. The antiviral activity of these compounds was tested against Junín virus (the etiological agent of Argentine hemorrhagic fever). The p-chlorophenyl derivatives showed antiviral activity in a range of micromolar concentration.


Assuntos
Antivirais/síntese química , Antivirais/farmacologia , Ribose/química , Tiadiazóis/síntese química , Tiadiazóis/farmacologia , Xilose/química , Antivirais/química , Técnicas de Química Sintética , Vírus Junin/efeitos dos fármacos , Tiadiazóis/química
6.
Bioorg Med Chem Lett ; 29(4): 556-559, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30612844

RESUMO

In this work, several ribavirin analogues were synthesized and incorporated into a multivalent arrangement. Both were subsequently modified by the addition of polyhydroxylated residues. Their antiviral activity was tested against Junín virus, etiological agent responsible of Argentine hemorrhagic fever. Some compounds inhibited Junín virus in the range of 13.2-389.1 µM. Two modified ribavirin analogues presented an effective concentration comparable to ribavirin but with a higher selectivity index.


Assuntos
Antivirais/farmacologia , Vírus Junin/efeitos dos fármacos , Ribavirina/análogos & derivados , Células A549 , Animais , Chlorocebus aethiops , Humanos , Células Vero
7.
Antiviral Res ; 157: 140-150, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30031760

RESUMO

Infection with Junín virus (JUNV) is currently being effectively managed in the endemic region using a combination of targeted vaccination and plasma therapy. However, the long-term sustainability of plasma therapy is unclear and similar resources are not available for other New World arenaviruses. As a result, there has been renewed interest regarding the potential of drug-based therapies. To facilitate work on this issue, we present the establishment and subsequent optimization of a JUNV minigenome system to a degree suitable for high-throughput miniaturization, thereby providing a screening platform focused solely on factors affecting RNA synthesis. Using this tool, we conducted a limited drug library screen and identified AVN-944, a non-competitive inosine monophosphate dehydrogenase (IMPDH) inhibitor, as an inhibitor of arenavirus RNA synthesis. We further developed a transcription and replication competent virus-like particle (trVLP) system based on these minigenomes and used it to screen siRNAs against IMPDH, verifying its role in supporting arenavirus RNA synthesis. The antiviral effect of AVN-944, as well as siRNA inhibition, on JUNV RNA synthesis supports that, despite playing only a minor role in the activity of ribavirin, exclusive IMPDH inhibitors may indeed have significant therapeutic potential for use against New World arenaviruses. Finally, we confirmed that AVN-944 is also active against arenavirus infection in cell culture, supporting the suitability of arenavirus lifecycle modelling systems as tools for the screening and identification, as well as the mechanistic characterization, of novel antiviral compounds.


Assuntos
Antivirais/isolamento & purificação , Carbamatos/isolamento & purificação , Inibidores Enzimáticos/isolamento & purificação , IMP Desidrogenase/metabolismo , Vírus Junin/efeitos dos fármacos , Vírus Junin/crescimento & desenvolvimento , Compostos de Fenilureia/isolamento & purificação , Animais , Antivirais/farmacologia , Carbamatos/farmacologia , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores Enzimáticos/farmacologia , Humanos , IMP Desidrogenase/antagonistas & inibidores , Vírus Junin/genética , Compostos de Fenilureia/farmacologia , Genética Reversa/métodos , Transcrição Gênica/efeitos dos fármacos , Cultura de Vírus , Replicação Viral/efeitos dos fármacos
8.
J Med Virol ; 90(5): 819-827, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29315647

RESUMO

The aim of this study was to investigate the effect of A771726, the active metabolite of leflunomide, (CONICET-UBA), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad against the infection with Junín virus (JUNV), agent of Argentine hemorrhagic fever (AHF). The treatment with non-cytotoxic concentrations of A771726 of Vero and A549 cells infected with JUNV inhibited virus replication in a dose-dependent manner, as determined by virus yield reduction assay. The antiviral effectiveness of A771726 was not importantly affected by the multiplicity of infection and the virus strain. Moreover, the combination of A771726 and ribavirin had a significantly more potent antiviral activity than each single drug treatment. Mechanistic studies showed that the main action of A771726 is exerted before 6 h of JUNV infection. Accordingly, inhibition of viral RNA synthesis was detected in treated infected cells by real time RT-PCR. The exogenous addition of uridine or orotic acid produced a partial reversal of the inhibitory effect of A771726 on infective virus production whereas a total reversion was detected on JUNV RNA synthesis, probably by restoration of the enzymatic activity of dihydroorotate dehydrogenase (DHODH) and the intracellular pyrimidine pools. In conclusion, these results suggest that the antiviral target would be viral RNA synthesis through pyrimidine depletion, but any other effect of the compound on JUNV infection cannot be excluded. This study opens the possibility of the therapeutic application of a wide spectrum host-targeted compound alone or in combination with ribavirin to combat AHF as well as other human pathogenic arenaviruses.


Assuntos
Compostos de Anilina/farmacologia , Antivirais/farmacologia , Hidroxibutiratos/farmacologia , Vírus Junin/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Células A549 , Animais , Chlorocebus aethiops , Crotonatos , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Humanos , Nitrilas , RNA Viral/biossíntese , Ribavirina/farmacologia , Toluidinas , Células Vero , Carga Viral
9.
Antiviral Res ; 145: 131-135, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28780425

RESUMO

A collection of Old and New World arenaviruses are etiologic agents of viral hemorrhagic fever, a syndrome that features hematologic abnormalities, vascular leak, hypovolemia, and multi-organ failure. Treatment is limited to ribavirin for Lassa fever and immune plasma for Argentine hemorrhagic fever. Improved therapeutic options that are safe, more effective and widely available are needed. Here, we show that modification of favipiravir treatment to include a high-dose loading period achieves complete protection in a guinea pig model of Argentine hemorrhagic fever when treatment was initiated two days following challenge with Junin virus (JUNV). This loading dose strategy also protected 50% of animals from lethal disease when treatment was delayed until 5 days post-infection and extended the survival time in those that succumbed. Consistent with the survival data, dramatic reductions in serum and tissue virus loads were observed in animals treated with favipiravir. This is the first report demonstrating complete protection against uniformly lethal JUNV infection in guinea pigs by administration of a small molecule antiviral drug.


Assuntos
Amidas/administração & dosagem , Antivirais/administração & dosagem , Febre Hemorrágica Americana/tratamento farmacológico , Vírus Junin/efeitos dos fármacos , Pirazinas/administração & dosagem , Amidas/uso terapêutico , Animais , Antivirais/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Cobaias , Febre Hemorrágica Americana/sangue , Febre Hemorrágica Americana/mortalidade , Pirazinas/uso terapêutico , Análise de Sobrevida , Carga Viral/efeitos dos fármacos
10.
J Virol Methods ; 246: 51-57, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28359770

RESUMO

Studies were conducted to determine the performance of four dyes in assessing antiviral activities of compounds against three RNA viruses with differing cytopathogenic properties. Dyes included alamarBlue® measured by absorbance (ALB-A) and fluorescence (ALB-F), neutral red (NR), Viral ToxGlo™ (VTG), and WST-1. Viruses were chikungunya, dengue type 2, and Junin, which generally cause 100, 80-90, and 50% maximal cytopathic effect (CPE), respectively, in Vero or Vero 76 cells Compounds evaluated were 6-azauridine, BCX-4430, 3-deazaguanine, EICAR, favipiravir, infergen, mycophenolic acid (MPA), ribavirin, and tiazofurin. The 50% virus-inhibitory (EC50) values for each inhibitor and virus combination did not vary significantly based on the dye used. However, dyes varied in distinguishing the vitality of virus-infected cultures when not all cells were killed by virus infection. For example, VTG uptake into dengue-infected cells was nearly 50% when visual examination showed only 10-20% cell survival. ALB-A measured infected cell viability differently than ALB-F as follows: 16% versus 32% (dengue-infected), respectively, and 51% versus 72% (Junin-infected), respectively. Cytotoxicity (CC50) assays with dyes in uninfected proliferating cells produced similar CC50 values for EICAR (1.5-8.9µM) and MPA (0.8-2.5µM). 6-Azauridine toxicity was 6.1-17.5µM with NR, VTG, and WST-1, compared to 48-92µM with ALB-A and ALB-F (P<0.001). Curiously, the CC50 values for 3-deazaguanine were 83-93µM with ALB-F versus 2.4-7.0µM with all other dyes including ALB-A (P<0.001). Overall, ALB minimized the toxicities detected with these two inhibitors. Because the choice of dyes affected CC50 values, this impacted on the resulting in vitro selectivity indexes (calculated as CC50/EC50 ratio).


Assuntos
Antivirais/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Corantes , Efeito Citopatogênico Viral , Vírus de RNA/efeitos dos fármacos , Vírus/efeitos dos fármacos , Animais , Vírus Chikungunya/efeitos dos fármacos , Vírus Chikungunya/patogenicidade , Vírus Chikungunya/fisiologia , Chlorocebus aethiops , Corantes/química , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/patogenicidade , Vírus da Dengue/fisiologia , Vírus Junin/efeitos dos fármacos , Vírus Junin/patogenicidade , Vírus Junin/fisiologia , Oxazinas , Vírus de RNA/patogenicidade , Vírus de RNA/fisiologia , Células Vero , Replicação Viral/efeitos dos fármacos , Xantenos
11.
J Virol ; 90(9): 4494-4510, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26912630

RESUMO

UNLABELLED: Virus entry into cells is a multistep process that often requires the subversion of subcellular machineries. A more complete understanding of these steps is necessary to develop new antiviral strategies. While studying the potential role of the actin network and one of its master regulators, the small GTPase Cdc42, during Junin virus (JUNV) entry, we serendipitously uncovered the small molecule ZCL278, reported to inhibit Cdc42 function as an entry inhibitor for JUNV and for vesicular stomatitis virus, lymphocytic choriomeningitis virus, and dengue virus but not for the nonenveloped poliovirus. Although ZCL278 did not interfere with JUNV attachment to the cell surface or virus particle internalization into host cells, it prevented the release of JUNV ribonucleoprotein cores into the cytosol and decreased pH-mediated viral fusion with host membranes. We also identified SVG-A astroglial cell-derived cells to be highly permissive for JUNV infection and generated new cell lines expressing fluorescently tagged Rab5c or Rab7a or lacking Cdc42 using clustered regularly interspaced short palindromic repeat (CRISPR)-caspase 9 (Cas9) gene-editing strategies. Aided by these tools, we uncovered that perturbations in the actin cytoskeleton or Cdc42 activity minimally affect JUNV entry, suggesting that the inhibitory effect of ZCL278 is not mediated by ZCL278 interfering with the activity of Cdc42. Instead, ZCL278 appears to redistribute viral particles from endosomal to lysosomal compartments. ZCL278 also inhibited JUNV replication in a mouse model, and no toxicity was detected. Together, our data suggest the unexpected antiviral activity of ZCL278 and highlight its potential for use in the development of valuable new tools to study the intracellular trafficking of pathogens. IMPORTANCE: The Junin virus is responsible for outbreaks of Argentine hemorrhagic fever in South America, where 5 million people are at risk. Limited options are currently available to treat infections by Junin virus or other viruses of the Arenaviridae, making the identification of additional tools, including small-molecule inhibitors, of great importance. How Junin virus enters cells is not yet fully understood. Here we describe new cell culture models in which the cells are susceptible to Junin virus infection and to which we applied CRISPR-Cas9 genome engineering strategies to help characterize early steps during virus entry. We also uncovered ZCL278 to be a new antiviral small molecule that potently inhibits the cellular entry of the Junin virus and other enveloped viruses. Moreover, we show that ZCL278 also functions in vivo, thereby preventing Junin virus replication in a mouse model, opening the possibility for the discovery of ZCL278 derivatives of therapeutic potential.


Assuntos
Antivirais/farmacologia , Benzamidas/farmacologia , Descoberta de Drogas , Tioureia/análogos & derivados , Internalização do Vírus/efeitos dos fármacos , Actinas/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Clatrina/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Endocitose/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Endossomos/virologia , Técnicas de Inativação de Genes , Febre Hemorrágica Americana/genética , Febre Hemorrágica Americana/metabolismo , Febre Hemorrágica Americana/virologia , Humanos , Vírus Junin/efeitos dos fármacos , Vírus Junin/fisiologia , Camundongos , Ligação Proteica , Transporte Proteico , Proteólise , Ribonucleoproteínas/metabolismo , Tioureia/farmacologia , Carga Viral , Proteínas Virais/metabolismo , Ligação Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo
12.
Antiviral Res ; 126: 62-8, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26711718

RESUMO

Favipiravir is approved in Japan to treat novel or re-emerging influenza viruses, and is active against a broad spectrum of RNA viruses, including Ebola. Ribavirin is the only other licensed drug with activity against multiple RNA viruses. Recent studies show that ribavirin and favipiravir act synergistically to inhibit bunyavirus infections in cultured cells and laboratory mice, likely due to their different mechanisms of action. Convalescent immune globulin is the only approved treatment for Argentine hemorrhagic fever caused by the rodent-borne Junin arenavirus. We previously reported that favipiravir is highly effective in a number of small animal models of Argentine hemorrhagic fever. We now report that addition of low dose of ribavirin synergistically potentiates the activity of favipiravir against Junin virus infection of guinea pigs and another arenavirus, Pichinde virus infection of hamsters. This suggests that the efficacy of favipiravir against hemorrhagic fever viruses can be further enhanced through the addition of low-dose ribavirin.


Assuntos
Amidas/farmacologia , Antivirais/farmacologia , Febres Hemorrágicas Virais/tratamento farmacológico , Pirazinas/farmacologia , Vírus de RNA/efeitos dos fármacos , Ribavirina/farmacologia , Animais , Arenavirus/efeitos dos fármacos , Chlorocebus aethiops , Cricetinae , Vírus da Dengue/efeitos dos fármacos , Modelos Animais de Doenças , Sinergismo Farmacológico , Feminino , Cobaias , Orthohantavírus/efeitos dos fármacos , Vírus da Febre Hemorrágica da Crimeia-Congo/efeitos dos fármacos , Febre Hemorrágica Americana/tratamento farmacológico , Doença pelo Vírus Ebola/tratamento farmacológico , Febres Hemorrágicas Virais/sangue , Febres Hemorrágicas Virais/veterinária , Febres Hemorrágicas Virais/virologia , Vírus Junin/efeitos dos fármacos , Masculino , Mesocricetus , Camundongos , Células Vero
13.
Arch Virol ; 160(2): 469-75, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25488290

RESUMO

In previous work, we demonstrated that the arenavirus Junín virus (JUNV) is able to activate Akt by means of the phosphatidylinositol-3-kinase (PI3K) survival pathway during virus entry. This work extends our study, emphasizing the relevance of this pathway in the establishment and maintenance of persistent infection in vitro. During the course of infection, JUNV-infected Vero cells showed a typical cytopathic effect that may be ascribed to apoptotic cell death. Treatment of infected cultures with Ly294002, an inhibitor of the PI3K/Akt pathway, produced an apoptotic response similar to that observed for uninfected cells treated with the drug. This result suggests that virus-induced activation of the PI3K/Akt pathway does not deliver a strong enough anti-apoptotic signal to explain the low proportion of apoptotic cells observed during infection. Also, inhibition of the PI3K/Akt pathway during the acute stage of infection did not prevent the establishment of persistence. Furthermore, treatment of persistently JUNV-infected cells with Ly294002 did not alter viral protein expression. These findings indicate that despite the positive modulation of the PI3/Akt pathway during Junín virus entry, this would not play a critical role in the establishment and maintenance of JUNV persistence in Vero cells.


Assuntos
Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Febre Hemorrágica Americana/virologia , Vírus Junin/efeitos dos fármacos , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Animais , Apoptose , Linhagem Celular , Chlorocebus aethiops , Febre Hemorrágica Americana/tratamento farmacológico , Vírus Junin/crescimento & desenvolvimento , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Células Vero , Proteínas Virais/biossíntese
14.
PLoS Negl Trop Dis ; 8(6): e2933, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24901990

RESUMO

The new world arenavirus Junín virus (JUNV) is the causative agent of Argentine hemorrhagic fever, a lethal human infectious disease. Adult laboratory mice are generally resistant to peripheral infection by JUNV. The mechanism underlying the mouse resistance to JUNV infection is largely unknown. We have reported that interferon receptor knockout mice succumb to JUNV infection, indicating the critical role of interferon in restricting JUNV infection in mice. Here we report that the pathogenic and vaccine strains of JUNV were highly sensitive to interferon in murine primary cells. Treatment with low concentrations of interferon abrogated viral NP protein expression in murine cells. The replication of both JUNVs was enhanced in IRF3/IRF7 deficient cells. In addition, the vaccine strain of JUNV displayed impaired growth in primary murine cells. Our data suggested a direct and potent role of host interferon response in restricting JUNV replication in mice. The defect in viral growth for vaccine JUNV might also partially explain its attenuation in mice.


Assuntos
Antivirais/farmacologia , Interferons/imunologia , Interferons/farmacologia , Vírus Junin/efeitos dos fármacos , Vírus Junin/imunologia , Animais , Células Cultivadas , Fator Regulador 3 de Interferon/deficiência , Fator Regulador 7 de Interferon/deficiência , Interferons/deficiência , Vírus Junin/crescimento & desenvolvimento , Vírus Junin/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Virais/biossíntese , Replicação Viral/efeitos dos fármacos
15.
Antivir Chem Chemother ; 23(4): 151-9, 2014 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-23337126

RESUMO

BACKGROUND: Junín virus (JUNV) and several other clade B New World arenaviruses cause human disease ranging from mild febrile illness to severe viral haemorrhagic fever. These viruses pose a significant threat to national security and safe and effective therapies are limited except in Argentina, where immune plasma is the standard of care for treating JUNV infection in cases of Argentine haemorrhagic fever. METHODS: An in vitro screen of the Chemtura library identified several compounds with activity against Tacaribe virus (TCRV), a clade B arenavirus closely related to JUNV. Of these compounds, D746, a phenolic dibenzylsulfide, was further pursued for additional in vitro studies and evaluated in the AG129 mouse TCRV infection model. RESULTS: D746 was found to act during an early to intermediate stage of the TCRV replication cycle and µM range activity was confirmed by virus yield reduction assays with both TCRV and JUNV. Although intraperitoneal twice daily treatment regimens were found to be highly effective when started 2 h prior to TCRV challenge in AG129 mice, post-exposure treatment initiated 3 days after infection was not efficacious. Interestingly, despite the pre-exposure treatment success, D746 did not reduce serum or tissue virus titres during the acute infection. Moreover, D746 elicited ascites fluid accumulation in mice during, as well as independent of, infection. CONCLUSIONS: Our findings suggest that D746 may be altering the host response to TCRV infection in AG129 mice in a way that limits pathogenesis and thereby protects mice from otherwise lethal infection in the absence of measurable reductions in viral burden.


Assuntos
Antivirais/química , Antivirais/uso terapêutico , Arenavirus do Novo Mundo/efeitos dos fármacos , Febre Hemorrágica Americana/tratamento farmacológico , Animais , Chlorocebus aethiops , Humanos , Vírus Junin/efeitos dos fármacos , Camundongos , Fenóis/química , Fenóis/uso terapêutico , Sulfetos/química , Sulfetos/uso terapêutico , Células Vero
16.
Sci Transl Med ; 5(204): 204ra131, 2013 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-24068738

RESUMO

New World hemorrhagic fever arenavirus infection results in 15 to 30% mortality in humans. We performed a high-throughput small interfering RNA screen with Junín virus glycoprotein-pseudotyped viruses to find potential host therapeutic targets. Voltage-gated calcium channel (VGCC) subunits, for which there are Food and Drug Administration (FDA)-approved drugs, were identified in the screen. Knockdown of VGCC subunits or treatment with channel blockers diminished Junín virus-cell fusion and entry into cells and thereby decreased infection. Gabapentin, an FDA-approved drug used to treat neuropathic pain that targets the α2δ2 subunit, inhibited infection of mice by the Candid 1 vaccine strain of the virus. These findings demonstrate that VGCCs play a role in virus infection and have the potential to lead to therapeutic intervention of New World arenavirus infection.


Assuntos
Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Ensaios de Triagem em Larga Escala , Vírus Junin/fisiologia , Terapia de Alvo Molecular , RNA Interferente Pequeno/genética , Internalização do Vírus , Aminas/farmacologia , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Fusão Celular , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Chlorocebus aethiops , Ácidos Cicloexanocarboxílicos/farmacologia , Gabapentina , Técnicas de Silenciamento de Genes , Interações Hospedeiro-Patógeno/genética , Humanos , Vírus Junin/efeitos dos fármacos , Camundongos , Ligação Proteica/efeitos dos fármacos , Subunidades Proteicas/metabolismo , Interferência de RNA , Receptores da Transferrina/metabolismo , Células Vero , Internalização do Vírus/efeitos dos fármacos , Ácido gama-Aminobutírico/farmacologia
17.
PLoS Negl Trop Dis ; 7(12): e2614, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24386500

RESUMO

BACKGROUND: Junín virus (JUNV), the etiologic agent of Argentine hemorrhagic fever (AHF), is classified by the NIAID and CDC as a Category A priority pathogen. Presently, antiviral therapy for AHF is limited to immune plasma, which is readily available only in the endemic regions of Argentina. T-705 (favipiravir) is a broadly active small molecule RNA-dependent RNA polymerase inhibitor presently in clinical evaluation for the treatment of influenza. We have previously reported on the in vitro activity of favipiravir against several strains of JUNV and other pathogenic New World arenaviruses. METHODOLOGY/PRINCIPAL FINDINGS: To evaluate the efficacy of favipiravir in vivo, guinea pigs were challenged with the pathogenic Romero strain of JUNV, and then treated twice daily for two weeks with oral or intraperitoneal (i.p.) favipiravir (300 mg/kg/day) starting 1-2 days post-infection. Although only 20% of animals treated orally with favipiravir survived the lethal challenge dose, those that succumbed survived considerably longer than guinea pigs treated with placebo. Consistent with pharmacokinetic analysis that showed greater plasma levels of favipiravir in animals dosed by i.p. injection, i.p. treatment resulted in a substantially higher level of protection (78% survival). Survival in guinea pigs treated with ribavirin was in the range of 33-40%. Favipiravir treatment resulted in undetectable levels of serum and tissue viral titers and prevented the prominent thrombocytopenia and leucopenia observed in placebo-treated animals during the acute phase of infection. CONCLUSIONS/SIGNIFICANCE: The remarkable protection afforded by i.p. favipiravir intervention beginning 2 days after challenge is the highest ever reported for a small molecule antiviral in the difficult to treat guinea pig JUNV challenge model. These findings support the continued development of favipiravir as a promising antiviral against JUNV and other related arenaviruses.


Assuntos
Amidas/uso terapêutico , Antivirais/uso terapêutico , Febre Hemorrágica Americana/tratamento farmacológico , Vírus Junin/efeitos dos fármacos , Pirazinas/uso terapêutico , Administração Oral , Amidas/farmacocinética , Animais , Antivirais/farmacocinética , Modelos Animais de Doenças , Cobaias , Febre Hemorrágica Americana/virologia , Injeções Intraperitoneais , Masculino , Plasma/química , Pirazinas/farmacocinética , Análise de Sobrevida , Viremia/prevenção & controle
18.
Biochem Biophys Res Commun ; 422(4): 590-5, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22595457

RESUMO

It has been previously described that S-layer binds to the C-type lectin DC-specific ICAM-3-grabbing nonintegrin (DC-SIGN, CD209). It was also shown that DC-SIGN is a cell-surface adhesion factor that enhances viral entry of several virus families. Among those, Junin virus (JUNV) entry is enhanced in cells expressing DC-SIGN and for that reason surface-layer protein (S-layer) of Lactobacillus acidophilus ATCC 4365 was evaluated as a possible JUNV inhibitor. Experiments using 3T3 cells stably expressing DC-SIGN, showed an almost complete inhibition of JUNV infection when they were treated with S-layer in a similar extend as the inhibition shown by mannan. However no inhibition effect was observed in 3T3 wild type cells or in 3T3 cells expressing liver/lymph node-specific ICAM-3 grabbing nonintegrin (L-SIGN or DC-SIGNR or CD209L). Treatments with S-layer during different times in the infection demonstrated that inhibition was only observed when S-layer was presented in early stages of the viral infection. This inhibition does not involve the classic recognition of mannose by this C-type lectin as the S-layer showed no evidence to be glycosylated. In fact, the highly basic nature of the S-layer (pI>9.5) seems to be involved in electrostatic interactions between DC-SIGN and S-layer, since high pH abolished the inhibitory effect on infection cause by the S-layer. In silico analysis predicts a Ca(2+)-dependant carbohydrate recognition domain in the SlpA protein. This novel characteristic of the S-layer, a GRAS status protein, contribute to the pathogen exclusion reported for this probiotic strain and may be applied as an antiviral agent to inhibit several kinds of viruses.


Assuntos
Antivirais/farmacologia , Proteínas de Bactérias/farmacologia , Vírus Junin/efeitos dos fármacos , Lactobacillus acidophilus , Glicoproteínas de Membrana/farmacologia , Internalização do Vírus/efeitos dos fármacos , Células 3T3 , Sequência de Aminoácidos , Animais , Antivirais/química , Antivirais/isolamento & purificação , Infecções por Arenaviridae , Proteínas de Bactérias/isolamento & purificação , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Chlorocebus aethiops , Sequência Consenso , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/isolamento & purificação , Camundongos , Dados de Sequência Molecular , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Células Vero
19.
Zoonoses Public Health ; 59(4): 278-85, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22212688

RESUMO

Junin virus (JUNV) is the aetiological agent of Argentine haemorrhagic fever. The pathogenesis of the infection is not well understood, no licensed vaccines exist and no specific antiviral therapy is available. Previous studies have demonstrated the ability of ribavirin to delay and reduce JUNV disease and virus burden in guinea pigs without preventing death. Based on available data, we performed three different studies to determine the efficacy of ribavirin against JUNV in the guinea pig model with a focus on survival. Different doses and treatment schedules of ribavirin were tested in a lethal model of JUNV infection. Our results show that prolonged treatment with high doses of ribavirin significantly reduces the mortality in guinea pigs infected with JUNV. These results may be useful in future experimental studies or clinical testing.


Assuntos
Antivirais/uso terapêutico , Infecções por Arenaviridae/tratamento farmacológico , Vírus Junin/efeitos dos fármacos , Ribavirina/uso terapêutico , Animais , Infecções por Arenaviridae/diagnóstico , Infecções por Arenaviridae/mortalidade , Infecções por Arenaviridae/virologia , Chlorocebus aethiops , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Cobaias , Vírus Junin/isolamento & purificação , Estimativa de Kaplan-Meier , Profilaxia Pós-Exposição , Células Vero
20.
Antiviral Res ; 93(1): 16-22, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22027649

RESUMO

There are no specific approved drugs for the treatment of agents of viral hemorrhagic fevers (HF) and antiviral therapies against these viruses are urgently needed. The present study characterizes the potent and selective antiviral activity against the HF causing arenavirus Junin virus (JUNV) of the compound 10-allyl-6-chloro-4-methoxy-9(10H)-acridone, designated 3f. The effectiveness of 3f to inhibit JUNV multiplication was not importantly affected by the initial multiplicity of infection, with similar effective concentration 50% (EC(50)) values in virus yield inhibition assays performed in Vero cells in the range of 0.2-40 plaque forming units (PFU)/cell. Mechanistic studies demonstrated that 3f did not affect the initial steps of adsorption and internalization. The subsequent process of viral RNA synthesis was strongly inhibited, as quantified by real time RT-PCR in compound-treated cells relative to non-treated cells. The addition of exogenous guanosine rescued the infectivity and RNA synthesis of JUNV in 3f-treated cells in a dose-dependent manner, but the reversal was partial, suggesting that the reduction of the GTP pool contributed to the antiviral activity of 3f, but it was not the main operative mechanism. The comparison of 3f with two other viral RNA inhibitors, ribavirin and mycophenolic acid, showed that ribavirin did not act against JUNV through the cellular enzyme inosine monophosphate dehydrogenase (IMPDH) inhibition whereas the anti-JUNV activity of mycophenolic acid was mainly targeted at this enzyme.


Assuntos
Acridonas/farmacologia , Compostos Alílicos/farmacologia , Antivirais/farmacologia , Vírus Junin/efeitos dos fármacos , RNA Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Acridonas/química , Compostos Alílicos/química , Animais , Antivirais/química , Chlorocebus aethiops , Efeito Citopatogênico Viral/efeitos dos fármacos , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Guanosina/farmacologia , Vírus Junin/genética , Testes de Sensibilidade Microbiana , RNA Viral/biossíntese , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...