Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Virol ; 98(7): e0071424, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-38809021

RESUMO

Lassa virus (LASV) is the causative agent of human Lassa fever which in severe cases manifests as hemorrhagic fever leading to thousands of deaths annually. However, no approved vaccines or antiviral drugs are currently available. Recently, we screened approximately 2,500 compounds using a recombinant vesicular stomatitis virus (VSV) expressing LASV glycoprotein GP (VSV-LASVGP) and identified a P-glycoprotein inhibitor as a potential LASV entry inhibitor. Here, we show that another identified candidate, hexestrol (HES), an estrogen receptor agonist, is also a LASV entry inhibitor. HES inhibited VSV-LASVGP replication with a 50% inhibitory concentration (IC50) of 0.63 µM. Importantly, HES also inhibited authentic LASV replication with IC50 values of 0.31 µM-0.61 µM. Time-of-addition and cell-based membrane fusion assays suggested that HES inhibits the membrane fusion step during virus entry. Alternative estrogen receptor agonists did not inhibit VSV-LASVGP replication, suggesting that the estrogen receptor itself is unlikely to be involved in the antiviral activity of HES. Generation of a HES-resistant mutant revealed that the phenylalanine at amino acid position 446 (F446) of LASVGP, which is located in the transmembrane region, conferred resistance to HES. Although mutation of F446 enhanced the membrane fusion activity of LASVGP, it exhibited reduced VSV-LASVGP replication, most likely due to the instability of the pre-fusion state of LASVGP. Collectively, our results demonstrated that HES is a promising anti-LASV drug that acts by inhibiting the membrane fusion step of LASV entry. This study also highlights the importance of the LASVGP transmembrane region as a target for anti-LASV drugs.IMPORTANCELassa virus (LASV), the causative agent of Lassa fever, is the most devastating mammarenavirus with respect to its impact on public health in West Africa. However, no approved antiviral drugs or vaccines are currently available. Here, we identified hexestrol (HES), an estrogen receptor agonist, as the potential antiviral candidate drug. We showed that the estrogen receptor itself is not involved in the antiviral activity. HES directly bound to LASVGP and blocked membrane fusion, thereby inhibiting LASV infection. Through the generation of a HES-resistant virus, we found that phenylalanine at position 446 (F446) within the LASVGP transmembrane region plays a crucial role in the antiviral activity of HES. The mutation at F446 caused reduced virus replication, likely due to the instability of the pre-fusion state of LASVGP. These findings highlight the potential of HES as a promising candidate for the development of antiviral compounds targeting LASV.


Assuntos
Antivirais , Febre Lassa , Vírus Lassa , Internalização do Vírus , Replicação Viral , Vírus Lassa/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Humanos , Antivirais/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Chlorocebus aethiops , Febre Lassa/virologia , Febre Lassa/tratamento farmacológico , Células Vero , Receptores de Estrogênio/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/genética , Linhagem Celular , Fenilalanina/farmacologia , Fenilalanina/análogos & derivados
2.
Proc Natl Acad Sci U S A ; 119(30): e2201208119, 2022 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-35858434

RESUMO

Completion of the Lassa virus (LASV) life cycle critically depends on the activities of the virally encoded, RNA-dependent RNA polymerase in replication and transcription of the viral RNA genome in the cytoplasm of infected cells. The contribution of cellular proteins to these processes remains unclear. Here, we applied proximity proteomics to define the interactome of LASV polymerase in cells under conditions that recreate LASV RNA synthesis. We engineered a LASV polymerase-biotin ligase (TurboID) fusion protein that retained polymerase activity and successfully biotinylated the proximal proteome, which allowed the identification of 42 high-confidence LASV polymerase interactors. We subsequently performed a small interfering RNA (siRNA) screen to identify those interactors that have functional roles in authentic LASV infection. As proof of principle, we characterized eukaryotic peptide chain release factor subunit 3a (eRF3a/GSPT1), which we found to be a proviral factor that physically associates with LASV polymerase. Targeted degradation of GSPT1 by a small-molecule drug candidate, CC-90009, resulted in strong inhibition of LASV infection in cultured cells. Our work demonstrates the feasibility of using proximity proteomics to illuminate and characterize yet-to-be-defined host-pathogen interactome, which can reveal new biology and uncover novel targets for the development of antivirals against highly pathogenic RNA viruses.


Assuntos
Acetamidas , Antivirais , Isoindóis , Vírus Lassa , Fatores de Terminação de Peptídeos , Piperidonas , RNA Polimerase Dependente de RNA , Proteínas Virais , Acetamidas/farmacologia , Acetamidas/uso terapêutico , Antivirais/farmacologia , Antivirais/uso terapêutico , Linhagem Celular Tumoral , Humanos , Isoindóis/farmacologia , Isoindóis/uso terapêutico , Febre Lassa/tratamento farmacológico , Vírus Lassa/efeitos dos fármacos , Fatores de Terminação de Peptídeos/metabolismo , Piperidonas/metabolismo , Piperidonas/farmacologia , Piperidonas/uso terapêutico , Mapas de Interação de Proteínas/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Proteoma , Proteômica , RNA Polimerase Dependente de RNA/metabolismo , Proteínas Virais/metabolismo
3.
PLoS Comput Biol ; 17(1): e1008535, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33411731

RESUMO

Lassa fever is an haemorrhagic fever caused by Lassa virus (LASV). There is no vaccine approved against LASV and the only recommended antiviral treatment relies on ribavirin, despite limited evidence of efficacy. Recently, the nucleotide analogue favipiravir showed a high antiviral efficacy, with 100% survival obtained in an otherwise fully lethal non-human primate (NHP) model of Lassa fever. However the mechanism of action of the drug is not known and the absence of pharmacokinetic data limits the translation of these results to the human setting. Here we aimed to better understand the antiviral effect of favipiravir by developping the first mathematical model recapitulating Lassa viral dynamics and treatment. We analyzed the viral dynamics in 24 NHPs left untreated or treated with ribavirin or favipiravir, and we put the results in perspective with those obtained with the same drugs in the context of Ebola infection. Our model estimates favipiravir EC50 in vivo to 2.89 µg.mL-1, which is much lower than what was found against Ebola virus. The main mechanism of action of favipiravir was to decrease virus infectivity, with an efficacy of 91% at the highest dose. Based on our knowledge acquired on the drug pharmacokinetics in humans, our model predicts that favipiravir doses larger than 1200 mg twice a day should have the capability to strongly reduce the production infectious virus and provide a milestone towards a future use in humans.


Assuntos
Amidas , Antivirais , Febre Lassa/virologia , Vírus Lassa , Pirazinas , Ribavirina , Amidas/farmacocinética , Amidas/farmacologia , Amidas/uso terapêutico , Animais , Antivirais/farmacocinética , Antivirais/farmacologia , Antivirais/uso terapêutico , Feminino , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Febre Lassa/tratamento farmacológico , Vírus Lassa/efeitos dos fármacos , Vírus Lassa/patogenicidade , Vírus Lassa/fisiologia , Macaca fascicularis , Modelos Biológicos , Pirazinas/farmacocinética , Pirazinas/farmacologia , Pirazinas/uso terapêutico , Ribavirina/farmacocinética , Ribavirina/farmacologia , Ribavirina/uso terapêutico , Carga Viral/efeitos dos fármacos
4.
Bioorg Chem ; 107: 104619, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33450541

RESUMO

Severe emerging and re-emerging viral infections such as Lassa fever, Avian influenza (AI), and COVID-19 caused by SARS-CoV-2 urgently call for new strategies for the development of broad-spectrum antivirals targeting conserved components in the virus life cycle. Viral lipids are essential components, and viral-cell membrane fusion is the required entry step for most unrelated enveloped viruses. In this paper, we identified a porphyrin derivative of protoporphyrin IX (PPIX) that showed broad antiviral activities in vitro against a panel of enveloped pathogenic viruses including Lassa virus (LASV), Machupo virus (MACV), and SARS-CoV-2 as well as various subtypes of influenza A viral strains with IC50 values ranging from 0.91 ± 0.25 µM to 1.88 ± 0.34 µM. A mechanistic study using influenza A/Puerto Rico/8/34 (H1N1) as a testing strain showed that PPIX inhibits the infection in the early stage of virus entry through biophysically interacting with the hydrophobic lipids of enveloped virions, thereby inhibiting the entry of enveloped viruses into host cells. In addition, the preliminary antiviral activities of PPIX were further assessed by testing mice infected with the influenza A/Puerto Rico/8/34 (H1N1) virus. The results showed that compared with the control group without drug treatment, the survival rate and mean survival time of the mice treated with PPIX were apparently prolonged. These data encourage us to conduct further investigations using PPIX as a lead compound for the rational design of lipid-targeting antivirals for the treatment of infection with enveloped viruses.


Assuntos
Antivirais/uso terapêutico , Infecções por Orthomyxoviridae/tratamento farmacológico , Protoporfirinas/uso terapêutico , Internalização do Vírus/efeitos dos fármacos , Animais , Antivirais/síntese química , Antivirais/metabolismo , Antivirais/farmacologia , Arenavirus do Novo Mundo/efeitos dos fármacos , Chlorocebus aethiops , Cães , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Vírus Lassa/efeitos dos fármacos , Células Madin Darby de Rim Canino , Masculino , Lipídeos de Membrana/metabolismo , Camundongos , Testes de Sensibilidade Microbiana , Protoporfirinas/síntese química , Protoporfirinas/metabolismo , Protoporfirinas/farmacologia , SARS-CoV-2/efeitos dos fármacos , Células Vero , Envelope Viral/efeitos dos fármacos
5.
J Virol ; 93(6)2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30626681

RESUMO

Arenaviruses are a large family of emerging enveloped negative-strand RNA viruses that include several causative agents of viral hemorrhagic fevers. For cell entry, human-pathogenic arenaviruses use different cellular receptors and endocytic pathways that converge at the level of acidified late endosomes, where the viral envelope glycoprotein mediates membrane fusion. Inhibitors of arenavirus entry hold promise for therapeutic antiviral intervention and the identification of "druggable" targets is of high priority. Using a recombinant vesicular stomatitis virus pseudotype platform, we identified the clotrimazole-derivative TRAM-34, a highly selective antagonist of the calcium-activated potassium channel KCa3.1, as a specific entry inhibitor for arenaviruses. TRAM-34 specifically blocked entry of most arenaviruses, including hemorrhagic fever viruses, but not Lassa virus and other enveloped viruses. Anti-arenaviral activity was likewise observed with the parental compound clotrimazole and the derivative senicapoc, whereas structurally unrelated KCa3.1 inhibitors showed no antiviral effect. Deletion of KCa3.1 by CRISPR/Cas9 technology did not affect the antiarenaviral effect of TRAM-34, indicating that the observed antiviral effect of clotrimazoles was independent of the known pharmacological target. The drug affected neither virus-cell attachment, nor endocytosis, suggesting an effect on later entry steps. Employing a quantitative cell-cell fusion assay that bypasses endocytosis, we demonstrate that TRAM-34 specifically inhibits arenavirus-mediated membrane fusion. In sum, we uncover a novel antiarenaviral action of clotrimazoles that currently undergo in vivo evaluation in the context of other human diseases. Their favorable in vivo toxicity profiles and stability opens the possibility to repurpose clotrimazole derivatives for therapeutic intervention against human-pathogenic arenaviruses.IMPORTANCE Emerging human-pathogenic arenaviruses are causative agents of severe hemorrhagic fevers with high mortality and represent serious public health problems. The current lack of a licensed vaccine and the limited treatment options makes the development of novel antiarenaviral therapeutics an urgent need. Using a recombinant pseudotype platform, we uncovered that clotrimazole drugs, in particular TRAM-34, specifically inhibit cell entry of a range of arenaviruses, including important emerging human pathogens, with the exception of Lassa virus. The antiviral effect was independent of the known pharmacological drug target and involved inhibition of the unusual membrane fusion mechanism of arenaviruses. TRAM-34 and its derivatives currently undergo evaluation against a number of human diseases and show favorable toxicity profiles and high stability in vivo Our study provides the basis for further evaluation of clotrimazole derivatives as antiviral drug candidates. Their advanced stage of drug development will facilitate repurposing for therapeutic intervention against human-pathogenic arenaviruses.


Assuntos
Antivirais/farmacologia , Arenavirus/efeitos dos fármacos , Clotrimazol/farmacologia , Fusão de Membrana/efeitos dos fármacos , Células A549 , Animais , Infecções por Arenaviridae/tratamento farmacológico , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Endocitose/efeitos dos fármacos , Células HEK293 , Células HeLa , Febres Hemorrágicas Virais/tratamento farmacológico , Febres Hemorrágicas Virais/virologia , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Vírus Lassa/efeitos dos fármacos , Células Vero , Proteínas do Envelope Viral/metabolismo , Ligação Viral/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos
6.
PLoS Pathog ; 14(12): e1007439, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30576397

RESUMO

Arenaviruses are a significant cause of hemorrhagic fever, an often-fatal disease for which there is no approved antiviral therapy. Lassa fever in particular generates high morbidity and mortality in West Africa, where the disease is endemic, and a recent outbreak in Nigeria was larger and more geographically diverse than usual. We are developing LHF-535, a small-molecule viral entry inhibitor that targets the arenavirus envelope glycoprotein, as a therapeutic candidate for Lassa fever and other hemorrhagic fevers of arenavirus origin. Using a lentiviral pseudotype infectivity assay, we determined that LHF-535 had sub-nanomolar potency against the viral envelope glycoproteins from all Lassa virus lineages, with the exception of the glycoprotein from the LP strain from lineage I, which was 100-fold less sensitive than that of other strains. This reduced sensitivity was mediated by a unique amino acid substitution, V434I, in the transmembrane domain of the envelope glycoprotein GP2 subunit. This position corresponds to the attenuation determinant of Candid#1, a live-attenuated Junín virus vaccine strain used to prevent Argentine hemorrhagic fever. Using a virus-yield reduction assay, we determined that LHF-535 potently inhibited Junín virus, but not Candid#1, and the Candid#1 attenuation determinant, F427I, regulated this difference in sensitivity. We also demonstrated that a daily oral dose of LHF-535 at 10 mg/kg protected mice from a lethal dose of Tacaribe virus. Serial passage of Tacaribe virus in LHF-535-treated Vero cells yielded viruses that were resistant to LHF-535, and the majority of drug-resistant viruses exhibited attenuated pathogenesis. These findings provide a framework for the clinical development of LHF-535 as a broad-spectrum inhibitor of arenavirus entry and provide an important context for monitoring the emergence of drug-resistant viruses.


Assuntos
Antivirais/farmacologia , Febre Lassa , Vírus Lassa/genética , Virulência/efeitos dos fármacos , Virulência/genética , Animais , Chlorocebus aethiops , Farmacorresistência Viral/efeitos dos fármacos , Farmacorresistência Viral/genética , Células HEK293 , Humanos , Vírus Lassa/efeitos dos fármacos , Camundongos , Mutação , Células Vero , Proteínas do Envelope Viral/genética
7.
PLoS Pathog ; 14(9): e1007322, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30265711

RESUMO

Lassa fever virus (LASV) is endemic in West Africa and causes severe hemorrhagic fever and sensorineural hearing loss. We identified a small molecule inhibitor of LASV and used it to analyze the mechanism of entry. Using a photo-reactive analog that retains antiviral activity as a probe, we identified the inhibitor target as lysosome-associated membrane protein 1 (LAMP1), a host factor that binds to the LASV glycoprotein (GP) during infection. We found that LAMP1 binding to LASV GP is cholesterol-dependent, and that the inhibitor blocks infection by competing with cholesterol in LAMP1. Mutational analysis of a docking-based model identified a putative inhibitor binding site in the cholesterol-binding pocket within the LAMP1 domain that binds GP. These findings identify a critical role for cholesterol in LASV entry and a potential target for therapeutic intervention.


Assuntos
Colesterol/metabolismo , Vírus Lassa/fisiologia , Vírus Lassa/patogenicidade , Proteínas de Membrana Lisossomal/fisiologia , Receptores Virais/fisiologia , Adamantano/análogos & derivados , Adamantano/química , Adamantano/farmacologia , Animais , Antivirais/química , Antivirais/farmacologia , Linhagem Celular , Chlorocebus aethiops , Células HEK293 , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Febre Lassa/etiologia , Vírus Lassa/efeitos dos fármacos , Proteínas de Membrana Lisossomal/antagonistas & inibidores , Proteínas de Membrana Lisossomal/genética , Modelos Moleculares , Mutação , Estabilidade Proteica , Estrutura Terciária de Proteína , Receptores Virais/antagonistas & inibidores , Receptores Virais/genética , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/fisiologia , Internalização do Vírus/efeitos dos fármacos
8.
J Virol ; 92(16)2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29899092

RESUMO

Lassa virus (LASV) belongs to the Mammarenavirus genus (family Arenaviridae) and causes severe hemorrhagic fever in humans. At present, there are no Food and Drug Administration (FDA)-approved drugs or vaccines specific for LASV. Here, high-throughput screening of an FDA-approved drug library was performed against LASV entry by using pseudotype virus bearing LASV envelope glycoprotein (GPC). Two hit compounds, lacidipine and phenothrin, were identified as LASV entry inhibitors in the micromolar range. A mechanistic study revealed that both compounds inhibited LASV entry by blocking low-pH-induced membrane fusion. Accordingly, lacidipine showed virucidal effects on the pseudotype virus of LASV. Adaptive mutant analyses demonstrated that replacement of T40, located in the ectodomain of the stable-signal peptide (SSP), with lysine (K) conferred LASV resistance to lacidipine. Furthermore, lacidipine showed antiviral activity against LASV, the closely related Mopeia virus (MOPV), and the New World arenavirus Guanarito virus (GTOV). Drug-resistant variants indicated that V36M in the ectodomain of the SSP mutant and V436A in the transmembrane domain of the GP2 mutant conferred GTOV resistance to lacidipine, suggesting the interface between SSP and GP2 is the target of lacidipine. This study shows that lacidipine is a candidate for LASV therapy, reinforcing the notion that the SSP-GP2 interface provides an entry-targeted platform for arenavirus inhibitor design.IMPORTANCE Currently, there is no approved therapy to treat Lassa fever; therefore, repurposing of approved drugs will accelerate the development of a therapeutic stratagem. In this study, we screened an FDA-approved library of drugs and identified two compounds, lacidipine and phenothrin, which inhibited Lassa virus entry by blocking low-pH-induced membrane fusion. Additionally, both compounds extended their inhibition against the entry of Guanarito virus, and the viral targets were identified as the SSP-GP2 interface.


Assuntos
Antivirais/farmacologia , Di-Hidropiridinas/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Ensaios de Triagem em Larga Escala/métodos , Vírus Lassa/efeitos dos fármacos , Piretrinas/farmacologia , Internalização do Vírus/efeitos dos fármacos , Arenaviridae/efeitos dos fármacos , Arenavirus do Novo Mundo/efeitos dos fármacos , Análise Mutacional de DNA , Farmacorresistência Viral , Vírus Lassa/fisiologia
9.
J Virol ; 92(5)2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29237830

RESUMO

Fatal infection with the highly pathogenic Lassa virus (LASV) is characterized by extensive viral dissemination, indicating broad tissue tropism. The major cellular receptor for LASV is the highly conserved extracellular matrix receptor dystroglycan (DG). Binding of LASV depends on DG's tissue-specific posttranslational modification with the unusual O-linked polysaccharide matriglycan. Interestingly, functional glycosylation of DG does not always correlate with viral tropism observed in vivo The broadly expressed phosphatidylserine (PS) receptors Axl and Tyro3 were recently identified as alternative LASV receptor candidates. However, their role in LASV entry is not entirely understood. Here, we examine LASV receptor candidates in primary human cells and found coexpression of Axl with differentially glycosylated DG. To study LASV receptor use in the context of productive arenavirus infection, we employed recombinant lymphocytic choriomeningitis virus expressing LASV glycoprotein (rLCMV-LASV GP) as a validated biosafety level 2 (BSL2) model. We confirm and extend previous work showing that Axl can contribute to LASV entry in the absence of functional DG using "apoptotic mimicry" in a way similar to that of other enveloped viruses. We further show that Axl-dependent LASV entry requires receptor activation and involves a pathway resembling macropinocytosis. Axl-mediated LASV entry is facilitated by heparan sulfate and critically depends on the late endosomal protein LAMP-1 as an intracellular entry factor. In endothelial cells expressing low levels of functional DG, both receptors are engaged by the virus and can contribute to productive entry. In sum, we characterize the role of Axl in LASV entry and provide a rationale for targeting Axl in antiviral therapy.IMPORTANCE The highly pathogenic arenavirus Lassa virus (LASV) represents a serious public health problem in Africa. Although the principal LASV receptor, dystroglycan (DG), is ubiquitously expressed, virus binding critically depends on DG's posttranslational modification, which does not always correlate with tissue tropism. The broadly expressed phosphatidylserine receptor Axl was recently identified as an alternative LASV receptor candidate, but its role in LASV entry is unclear. Here, we investigate the exact role of Axl in LASV entry as a function of DG's posttranslational modification. We found that in the absence of functional DG, Axl can mediate LASV entry via apoptotic mimicry. Productive entry requires virus-induced receptor activation, involves macropinocytosis, and critically depends on LAMP-1. In endothelial cells that express low levels of glycosylated DG, both receptors can promote LASV entry. In sum, our study defines the roles of Axl in LASV entry and provides a rationale for targeting Axl in antiviral therapy.


Assuntos
Distroglicanas/metabolismo , Vírus Lassa/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Receptores Virais/metabolismo , Ligação Viral , Internalização do Vírus , Células A549 , Antivirais/farmacologia , Infecções por Arenaviridae/metabolismo , Linhagem Celular Tumoral , Distroglicanas/genética , Endossomos/metabolismo , Expressão Gênica , Glicosilação , Células HEK293 , Células HeLa , Heparitina Sulfato/farmacologia , Humanos , Vírus Lassa/efeitos dos fármacos , Vírus Lassa/patogenicidade , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/metabolismo , Proteínas de Membrana Lisossomal/metabolismo , Pinocitose/fisiologia , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/genética , Interferência de RNA , Receptores Proteína Tirosina Quinases/efeitos dos fármacos , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais , Tropismo , Receptor Tirosina Quinase Axl
10.
J Antimicrob Chemother ; 69(8): 2123-31, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24710028

RESUMO

OBJECTIVES: Filoviruses such as Ebola virus and Marburg virus cause a severe haemorrhagic fever syndrome in humans for which there is no specific treatment. Since filoviruses use a complex route of cell entry that depends on numerous cellular factors, we hypothesized that there may be drugs already approved for human use for other indications that interfere with signal transduction or other cellular processes required for their entry and hence have anti-filoviral properties. METHODS: We used authentic filoviruses and lentiviral particles pseudotyped with filoviral glycoproteins to identify and characterize such compounds. RESULTS: We discovered that amiodarone, a multi-ion channel inhibitor and adrenoceptor antagonist, is a potent inhibitor of filovirus cell entry at concentrations that are routinely reached in human serum during anti-arrhythmic therapy. A similar effect was observed with the amiodarone-related agent dronedarone and the L-type calcium channel blocker verapamil. Inhibition by amiodarone was concentration dependent and similarly affected pseudoviruses as well as authentic filoviruses. Inhibition of filovirus entry was observed with most but not all cell types tested and was accentuated by the pre-treatment of cells, indicating a host cell-directed mechanism of action. The New World arenavirus Guanarito was also inhibited by amiodarone while the Old World arenavirus Lassa and members of the Rhabdoviridae (vesicular stomatitis virus) and Bunyaviridae (Hantaan) families were largely resistant. CONCLUSIONS: The ion channel blockers amiodarone, dronedarone and verapamil inhibit filoviral cell entry.


Assuntos
Ebolavirus/efeitos dos fármacos , Marburgvirus/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Internalização do Vírus/efeitos dos fármacos , Antagonistas Adrenérgicos/farmacologia , Amiodarona/análogos & derivados , Amiodarona/farmacologia , Animais , Arenavirus do Novo Mundo/efeitos dos fármacos , Bunyaviridae/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/farmacologia , Linhagem Celular , Dronedarona , Humanos , Vírus Lassa/efeitos dos fármacos , Verapamil/farmacologia , Vírus da Estomatite Vesicular Indiana/efeitos dos fármacos
11.
Antiviral Res ; 228: 105923, 2024 08.
Artigo em Inglês | MEDLINE | ID: mdl-38844175

RESUMO

There are no approved vaccines or therapeutics for Lassa virus (LASV) infections. To identify compounds with anti-LASV activity, we conducted a cell-based screening campaign at biosafety level 4 and tested almost 60,000 compounds for activity against an infectious reporter LASV. Hits from this screen included several structurally related macrocycles. The most potent, Mac128, had a sub-micromolar EC50 against the reporter virus, inhibited wild-type clade IV LASV, and reduced viral titers by 4 orders of magnitude. Mechanistic studies suggested that Mac128 inhibited viral replication at the level of the polymerase.


Assuntos
Antivirais , Vírus Lassa , Compostos Macrocíclicos , Replicação Viral , Vírus Lassa/efeitos dos fármacos , Antivirais/farmacologia , Antivirais/química , Replicação Viral/efeitos dos fármacos , Compostos Macrocíclicos/farmacologia , Compostos Macrocíclicos/química , Humanos , Animais , Chlorocebus aethiops , Células Vero , Febre Lassa/virologia , Febre Lassa/tratamento farmacológico , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo , Proteínas Virais/genética
12.
Viruses ; 16(9)2024 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-39339831

RESUMO

High-consequence pathogens such as the Ebola, Marburg, and Lassa viruses are handled in maximum-containment biosafety level 4 (BSL-4) laboratories. Genetic material is often isolated from such viruses and subsequently removed from BSL-4 laboratories for a multitude of downstream analyses using readily accessible technologies and equipment available at lower-biosafety level laboratories. However, it is essential to ensure that these materials are free of viable viruses before removal from BSL-4 laboratories to guarantee sample safety. This study details the in-house procedure used for validating the inactivation of Ebola, Marburg, and Lassa virus cultures after incubation with AVL lysis buffer (Qiagen) and ethanol. This study's findings show that no viable virus was detectable when high-titer cultures of Ebola, Marburg, and Lassa viruses were incubated with AVL lysis buffer for 10 min, followed by an equal volume of 95% ethanol for 3 min, using a method with a sensitivity of ≤0.8 log10 TCID50 as the limit of detection.


Assuntos
Ebolavirus , Etanol , Vírus Lassa , Marburgvirus , Inativação de Vírus , Vírus Lassa/efeitos dos fármacos , Marburgvirus/efeitos dos fármacos , Ebolavirus/efeitos dos fármacos , Ebolavirus/fisiologia , Etanol/farmacologia , Inativação de Vírus/efeitos dos fármacos , Animais , Humanos , Contenção de Riscos Biológicos/métodos , Febre Lassa/virologia , Cultura de Vírus/métodos , Chlorocebus aethiops , Células Vero
13.
SLAS Discov ; 29(6): 100178, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39159824

RESUMO

Emerging highly pathogenic viruses can pose profound impacts on global health, the economy, and society. To meet that challenge, the National Institute of Allergy and Infectious Diseases (NIAID) established nine Antiviral Drug Discovery (AViDD) centers for early-stage identification and validation of novel antiviral drug candidates against viruses with pandemic potential. As part of this initiative, we established paired entry assays that simultaneously screen for inhibitors specifically targeting SARS-CoV-2 (SARS2), Lassa virus (LASV) and Machupo virus (MACV) entry. To do so we employed a dual pseudotyped virus (PV) infection system allowing us to screen ∼650,000 compounds efficiently and cost-effectively. Adaptation of these paired assays into 1536 well-plate format for ultra-high throughput screening (uHTS) resulted in the largest screening ever conducted in our facility, with over 2.4 million wells completed. The paired infection system allowed us to detect two PV infections simultaneously: LASV + MACV, MACV + SARS2, and SARS2 + LASV. Each PV contains a different luciferase reporter gene which enabled us to measure the infection of each PV exclusively, albeit in the same well. Each PV was screened at least twice utilizing different reporters, which allowed us to select the inhibitors specific to a particular PV and to exclude those that hit off targets, including cellular components or the reporter proteins. All assays were robust with an average Z' value ranging from 0.5 to 0.8. The primary screening of ∼650,000 compounds resulted in 1812, 1506, and 2586 unique hits for LASV, MACV, and SARS2, respectively. The confirmation screening narrowed this list further to 60, 40, and 90 compounds that are unique to LASV, MACV, and SARS2, respectively. Of these compounds, 8, 35, and 50 compounds showed IC50 value < 10 µM, some of which have much greater potency and excellent antiviral activity profiles specific to LASV, MACV, and SARS2, and none are cytotoxic. These selected compounds are currently being studied for their mechanism of action and to improve their specificity and potency through chemical modification.


Assuntos
Antivirais , Ensaios de Triagem em Larga Escala , Vírus Lassa , SARS-CoV-2 , Internalização do Vírus , Humanos , SARS-CoV-2/efeitos dos fármacos , Antivirais/farmacologia , Internalização do Vírus/efeitos dos fármacos , Vírus Lassa/efeitos dos fármacos , Ensaios de Triagem em Larga Escala/métodos , COVID-19/virologia , Descoberta de Drogas/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Tratamento Farmacológico da COVID-19 , Chlorocebus aethiops , Animais
14.
J Virol ; 86(8): 4578-85, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22345463

RESUMO

Several arenaviruses, chiefly Lassa virus (LASV), cause hemorrhagic fever (HF) disease in humans and pose a significant public health concern in regions where they are endemic. On the other hand, evidence indicates that the globally distributed prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) is a neglected human pathogen. The phosphatidylinositol 3-kinase (PI3K)/Akt pathway participates in many cellular processes, including cell survival and differentiation, and also has been shown to play important roles in different steps of the life cycles of a variety of viruses. Here we report that the inhibition of the PI3K/Akt pathway inhibited budding and to a lesser extent RNA synthesis, but not cell entry, of LCMV. Accordingly, BEZ-235, a PI3K inhibitor currently in cancer clinical trials, inhibited LCMV multiplication in cultured cells. These findings, together with those previously reported for Junin virus (JUNV), indicate that targeting the PI3K/Akt pathway could represent a novel antiviral strategy to combat human-pathogenic arenaviruses.


Assuntos
Arenavirus/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Liberação de Vírus , Animais , Arenavirus/efeitos dos fármacos , Arenavirus/genética , Linhagem Celular , Chlorocebus aethiops , Cromonas/farmacologia , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Humanos , Imidazóis/farmacologia , Vírus Lassa/efeitos dos fármacos , Vírus Lassa/genética , Vírus Lassa/metabolismo , Vírus da Coriomeningite Linfocítica/efeitos dos fármacos , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/metabolismo , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Quinolinas/farmacologia , RNA Viral/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Internalização do Vírus/efeitos dos fármacos , Liberação de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
15.
Bioorg Med Chem Lett ; 23(21): 5840-3, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24064500

RESUMO

Previously we reported the optimization of antiviral scaffolds containing benzimidazole and related heterocycles possessing activity against a variety of arenaviruses. These series of compounds were discovered through an HTS campaign of a 400,000 small molecule library using lentivirus-based pseudotypes incorporated with the Lassa virus envelope glycoprotein (LASV GP). This screening also uncovered an alternate series of very potent arenavirus inhibitors based upon an acylhydrazone scaffold. Subsequent SAR analysis of this chemical series involved various substitutions throughout the chemical framework along with assessment of the preferred stereochemistry. These studies led to an optimized analog (ST-161) possessing subnanomolar activity against LASV and submicromolar activity against a number of other viruses in the Arenaviridae family.


Assuntos
Antivirais/química , Antivirais/farmacologia , Hidrazonas/química , Hidrazonas/farmacologia , Vírus Lassa/efeitos dos fármacos , Acilação , Descoberta de Drogas , Humanos , Febre Lassa/tratamento farmacológico , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia
16.
Bioorg Med Chem Lett ; 23(3): 750-6, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23265900

RESUMO

A series of potent arenavirus inhibitors sharing a benzimidazole core were previously reported by our group. SAR studies were expanded beyond the previous analysis, which involved the attached phenyl rings and methylamino linker portion, to include modifications focused on the benzimidazole core. These changes included the introduction of various substituents to the bicyclic benzimidazole ring system along with alternate core heterocycles. Many of the analogs containing alternate nitrogen-based bicyclic ring systems were found to retain antiviral potency compared to the benzimidazole series from which we derived our lead compound, ST-193. In fact, 21 h, built on an imidazopyridine core, possessed a near tenfold increase in potency against Lassa virus pseudotypes compared to ST-193. As found with the benzimidazole series, broad-spectrum arenavirus activity was also observed for a number of the analogs discovered during this study.


Assuntos
Antivirais/síntese química , Antivirais/farmacologia , Arenavirus/efeitos dos fármacos , Benzimidazóis/química , Descoberta de Drogas , Compostos Heterocíclicos/síntese química , Antivirais/química , Benzimidazóis/síntese química , Benzimidazóis/farmacologia , Compostos Heterocíclicos/química , Compostos Heterocíclicos/farmacologia , Vírus Lassa/efeitos dos fármacos , Relação Estrutura-Atividade
17.
J Virol ; 85(2): 795-803, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21068251

RESUMO

Arenaviruses merit interest as clinically important human pathogens and include several causative agents, chiefly Lassa virus (LASV), of hemorrhagic fever disease in humans. There are no licensed LASV vaccines, and current antiarenavirus therapy is limited to the use of ribavirin, which is only partially effective and is associated with significant side effects. The arenavirus glycoprotein (GP) precursor GPC is processed by the cellular site 1 protease (S1P) to generate the peripheral virion attachment protein GP1 and the fusion-active transmembrane protein GP2, which is critical for production of infectious progeny and virus propagation. Therefore, S1P-mediated processing of arenavirus GPC is a promising target for therapeutic intervention. To this end, we have evaluated the antiarenaviral activity of PF-429242, a recently described small-molecule inhibitor of S1P. PF-429242 efficiently prevented the processing of GPC from the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) and LASV, which correlated with the compound's potent antiviral activity against LCMV and LASV in cultured cells. In contrast, a recombinant LCMV expressing a GPC whose processing into GP1 and GP2 was mediated by furin, instead of S1P, was highly resistant to PF-429242 treatment. PF-429242 did not affect virus RNA replication or budding but had a modest effect on virus cell entry, indicating that the antiarenaviral activity of PF-429242 was mostly related to its ability to inhibit S1P-mediated processing of arenavirus GPC. Our findings support the feasibility of using small-molecule inhibitors of S1P-mediated processing of arenavirus GPC as a novel antiviral strategy.


Assuntos
Antivirais/farmacologia , Vírus Lassa/efeitos dos fármacos , Vírus da Coriomeningite Linfocítica/efeitos dos fármacos , Pró-Proteína Convertases/antagonistas & inibidores , Inibidores de Proteases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas do Envelope Viral/metabolismo , Animais , Linhagem Celular , Humanos , Vírus Lassa/fisiologia , Vírus da Coriomeningite Linfocítica/fisiologia , Serina Endopeptidases , Internalização do Vírus/efeitos dos fármacos
18.
Arch Virol ; 157(1): 121-7, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21947546

RESUMO

Arenaviruses and filoviruses are capable of causing hemorrhagic fever syndrome in humans. Limited therapeutic and/or prophylactic options are available for humans suffering from viral hemorrhagic fever. In this report, we demonstrate that pre-treatment of host cells with the kinase inhibitors genistein and tyrphostin AG1478 leads to inhibition of infection or transduction in cells infected with Ebola virus, Marburg virus, and Lassa virus. In all, the results demonstrate that a kinase inhibitor cocktail consisting of genistein and tyrphostin AG1478 is a broad-spectrum antiviral that may be used as a therapeutic or prophylactic against arenavirus and filovirus hemorrhagic fever.


Assuntos
Antivirais/farmacologia , Ebolavirus/efeitos dos fármacos , Genisteína/farmacologia , Doença pelo Vírus Ebola/virologia , Febre Lassa/virologia , Vírus Lassa/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Tirfostinas/farmacologia , Linhagem Celular , Ebolavirus/genética , Ebolavirus/fisiologia , Doença pelo Vírus Ebola/tratamento farmacológico , Humanos , Febre Lassa/tratamento farmacológico , Vírus Lassa/genética , Vírus Lassa/fisiologia
19.
PLoS Negl Trop Dis ; 16(1): e0010089, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34990453

RESUMO

BACKGROUND: Only one recommendation currently exists for the treatment of Lassa fever (LF), which is ribavirin administered in conjunction with supportive care. This recommendation is primarily based on evidence generated from a single clinical trial that was conducted more than 30 years ago-the methodology and results of which have recently come under scrutiny. The requirement for novel therapeutics and reassessment of ribavirin is therefore urgent. However, a significant amount of work now needs to be undertaken to ensure that future trials for LF can be conducted consistently and reliably to facilitate the efficient generation of evidence. METHODOLOGY: We convened a consultation group to establish the position of clinicians and researchers on the core components of future trials. A Core Eligibility Criteria (CEC), Core Case Definition (CCD), Core Outcome Set (COS) and Core Data Variables (CDV) were developed through the process of a multi-stakeholder consultation that took place using a modified-Delphi methodology. RESULTS: A consensus position was achieved for each aspect of the framework, which accounts for the inclusion of pregnant women and children in future LF clinical trials. The framework consists of 8 core criteria, as well as additional considerations for trial protocols. CONCLUSIONS: This project represents the first step towards delineating the clinical development pathway for new Lassa fever therapeutics, following a period of 40 years without advancement. Future planned projects will bolster the work initiated here to continue the advancement of LF clinical research through a regionally-centred, collaborative methodology, with the aim of delineating a clear pathway through which LF clinical trials can progress efficiently and ensure sustainable investments are made in research capacity at a regional level.


Assuntos
Antivirais/farmacologia , Ensaios Clínicos Fase III como Assunto/métodos , Desenvolvimento de Medicamentos/métodos , Febre Lassa/tratamento farmacológico , Descoberta de Drogas/métodos , Humanos , Vírus Lassa/efeitos dos fármacos , Projetos de Pesquisa , Inquéritos e Questionários
20.
Antiviral Res ; 197: 105230, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34965446

RESUMO

Lassa virus (LASV) belongs to the Old World genus Mammarenavirus, family Arenaviridae, and order Bunyavirales. Arenavirus contains a segmented negative-sense RNA genome, which is in line with the bunyavirus and orthomyxoviruses. The segmented negative-sense RNA viruses utilize a cap-snatching strategy to provide primers cleavaged from the host capped mRNA for viral mRNA transcription. As a similar strategy and the conformational conservation shared with these viruses, the endonuclease (EN) would serve as an attractive target for developing broad-spectrum inhibitors. Using the LASV minigenome (MG) system, we screened a fragment-based drug discovery library and found that two hits, F1204 and F1781, inhibited LASV MG activity. Both hits also inhibited the prototype arenavirus Lymphocytic choriomeningitis virus (LCMV) MG activity. Furthermore, both hits effectively inhibited authentic LCMV and severe fever with thrombocytopenia syndrome virus (SFTSV) infections. Similarly, both hits could inhibit the activity of LASV, LCMV, and SFTSV EN. The combination of either compound with an arenavirus entry inhibitor had significant synergistic antiviral effects. Moreover, both hits were found to be capable of binding to LASV EN with a binding affinity at the micromolar level. These findings provide a basis for developing the hits as potential candidates for the treatment of segmented negative-sense RNA virus infections.


Assuntos
Antivirais/farmacologia , Descoberta de Drogas/métodos , Endonucleases/antagonistas & inibidores , Vírus Lassa/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Internalização do Vírus/efeitos dos fármacos , Animais , Antivirais/isolamento & purificação , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Células HEK293 , Ensaios de Triagem em Larga Escala/métodos , Humanos , Febre Lassa/tratamento farmacológico , Vírus Lassa/enzimologia , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA