Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Expert Opin Drug Discov ; 19(5): 537-551, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38606475

RESUMO

INTRODUCTION: Mammarenaviruses are negative-sense bisegmented enveloped RNA viruses that are endemic in Africa, the Americas, and Europe. Several are highly virulent, causing acute human diseases associated with high case fatality rates, and are considered to be significant with respect to public health impact or bioterrorism threat. AREAS COVERED: This review summarizes the status quo of treatment development, starting with drugs that are in advanced stages of evaluation in early clinical trials, followed by promising candidate medical countermeasures emerging from bench analyses and investigational animal research. EXPERT OPINION: Specific therapeutic treatments for diseases caused by mammarenaviruses remain limited to the off-label use of ribavirin and transfusion of convalescent sera. Progress in identifying novel candidate medical countermeasures against mammarenavirus infection has been slow in part because of the biosafety and biosecurity requirements. However, novel methodologies and tools have enabled increasingly efficient high-throughput molecular screens of regulatory-agency-approved small-molecule drugs and led to the identification of several compounds that could be repurposed for the treatment of infection with several mammarenaviruses. Unfortunately, most of them have not yet been evaluated in vivo. The most promising treatment under development is a monoclonal antibody cocktail that is protective against multiple lineages of the Lassa virus in nonhuman primate disease models.


Assuntos
Antivirais , Infecções por Arenaviridae , Arenaviridae , Desenvolvimento de Medicamentos , Humanos , Animais , Antivirais/farmacologia , Infecções por Arenaviridae/tratamento farmacológico , Infecções por Arenaviridae/virologia , Arenaviridae/efeitos dos fármacos , Virulência , Desenho de Fármacos
2.
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
3.
J Virol ; 95(24): e0139921, 2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34586865

RESUMO

Targeting host factors is a promising strategy to develop broad-spectrum antiviral drugs. Drugs targeting anti-apoptotic Bcl-2 family proteins that were originally developed as tumor suppressors have been reported to inhibit multiplication of different types of viruses. However, the mechanisms whereby Bcl-2 inhibitors exert their antiviral activity remain poorly understood. In this study, we have investigated the mechanisms by which obatoclax (OLX) and ABT-737 Bcl-2 inhibitors exhibited a potent antiviral activity against the mammarenavirus lymphocytic choriomeningitis virus (LCMV). OLX and ABT-737 potent anti-LCMV activity was not associated with their proapoptotic properties but rather with their ability to induce cell arrest at the G0/G1 phase. OLX- and ABT-737-mediated inhibition of Bcl-2 correlated with reduced expression levels of thymidine kinase 1 (TK1), cyclin A2 (CCNA2), and cyclin B1 (CCNB1) cell cycle regulators. In addition, small interfering RNA (siRNA)-mediated knockdown of TK1, CCNA2, and CCNB1 resulted in reduced levels of LCMV multiplication. The antiviral activity exerted by Bcl-2 inhibitors correlated with reduced levels of viral RNA synthesis at early times of infection. Importantly, ABT-737 exhibited moderate efficacy in a mouse model of LCMV infection, and Bcl-2 inhibitors displayed broad-spectrum antiviral activities against different mammarenaviruses and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Our results suggest that Bcl-2 inhibitors, actively being explored as anticancer therapeutics, might be repositioned as broad-spectrum antivirals. IMPORTANCE Antiapoptotic Bcl-2 inhibitors have been shown to exert potent antiviral activities against various types of viruses via mechanisms that are currently poorly understood. This study has revealed that Bcl-2 inhibitors' mediation of cell cycle arrest at the G0/G1 phase, rather than their proapoptotic activity, plays a critical role in blocking mammarenavirus multiplication in cultured cells. In addition, we show that Bcl-2 inhibitor ABT-737 exhibited moderate antimammarenavirus activity in vivo and that Bcl-2 inhibitors displayed broad-spectrum antiviral activities against different mammarenaviruses and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Our results suggest that Bcl-2 inhibitors, actively being explored as anticancer therapeutics, might be repositioned as broad-spectrum antivirals.


Assuntos
Apoptose , Arenaviridae/efeitos dos fármacos , Tratamento Farmacológico da COVID-19 , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Células A549 , Animais , Antivirais/farmacologia , Proteínas Reguladoras de Apoptose/farmacologia , Compostos de Bifenilo/farmacologia , COVID-19/virologia , Ciclo Celular , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/virologia , Chlorocebus aethiops , Ciclina A2/biossíntese , Ciclina B1/biossíntese , Fase G1 , Humanos , Indóis/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Nitrofenóis/farmacologia , Piperazinas/farmacologia , Pirróis/farmacologia , Fase de Repouso do Ciclo Celular , SARS-CoV-2 , Sulfonamidas/farmacologia , Timidina Quinase/biossíntese , Células Vero
4.
Viruses ; 13(7)2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34206216

RESUMO

Mammarenaviruses are prevalent pathogens distributed worldwide, and several strains cause severe cases of human infections with high morbidity and significant mortality. Currently, there is no FDA-approved antiviral drugs and vaccines against mammarenavirus and the potential treatment option is limited to an off-label use of ribavirin that shows only partial protective effect and associates with side effects. For the past few decades, extensive research has reported potential anti-mammarenaviral drugs and their mechanisms of action in host as well as vaccine candidates. This review describes current knowledge about mammarenavirus virology, progress of antiviral drug development, and technical strategies of drug screening.


Assuntos
Antivirais/farmacologia , Arenaviridae/efeitos dos fármacos , Desenvolvimento de Medicamentos/métodos , Ensaios de Triagem em Larga Escala , Células A549 , Animais , Arenaviridae/patogenicidade , Chlorocebus aethiops , Ensaios Clínicos como Assunto , Reposicionamento de Medicamentos , Células HEK293 , Humanos , Ribavirina/farmacologia , Células Vero , Replicação Viral/efeitos dos fármacos
5.
Viruses ; 12(8)2020 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-32751087

RESUMO

Mammarenaviruses cause chronic infections in rodents, which are their predominant natural hosts. Human infection with some of these viruses causes high-consequence disease, posing significant issues in public health. Currently, no FDA-licensed mammarenavirus vaccines are available, and anti-mammarenavirus drugs are limited to an off-label use of ribavirin, which is only partially efficacious and associated with severe side effects. Dihydroorotate dehydrogenase (DHODH) inhibitors, which block de novo pyrimidine biosynthesis, have antiviral activity against viruses from different families, including Arenaviridae, the taxonomic home of mammarenaviruses. Here, we evaluate five novel DHODH inhibitors for their antiviral activity against mammarenaviruses. All tested DHODH inhibitors were potently active against lymphocytic choriomeningitis virus (LCMV) (half-maximal effective concentrations [EC50] in the low nanomolar range, selectivity index [SI] > 1000). The tested DHODH inhibitors did not affect virion cell entry or budding, but rather interfered with viral RNA synthesis. This interference resulted in a potent interferon-independent inhibition of mammarenavirus multiplication in vitro, including the highly virulent Lassa and Junín viruses.


Assuntos
Antivirais/farmacologia , Arenaviridae/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/antagonistas & inibidores , Animais , Arenaviridae/classificação , Arenaviridae/fisiologia , Chlorocebus aethiops , Di-Hidro-Orotato Desidrogenase , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/química , Células HEK293 , Humanos , Interferons , Camundongos , Camundongos Endogâmicos C57BL , Inibidores da Síntese de Ácido Nucleico/química , Inibidores da Síntese de Ácido Nucleico/farmacologia , Pirimidinas/biossíntese , Células Vero , Replicação Viral/efeitos dos fármacos
6.
Bioorg Med Chem Lett ; 29(19): 126613, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31431358

RESUMO

The potent antiviral properties of 3-deazaneplanocin, 3-deaza-isoneplanocins (1) and recently discovered l-like carbocyclic nucleosides (2, 3 and 4) prompted us to pursue rationally conceived l-like 3-deazaneplanocin analogues. The synthesis of those analogues including l-like 3-deazaneplanocin (5), l-like 3-bromo-3-deazaneplanocin (6), and l-like 5'-fluoro-5'-deoxy-3-deazaneplanocin (7), was accomplished from a common intermediate, (-)-cyclopentenone (8). Antiviral analysis found 5 and 6 to display favorable activity against the Ebola virus, as expected for 3-deazaadenine carbocyclic nucleosides. Compound 5 also showed activity against arenaviruses, including Pinchinde and Tacaribe.


Assuntos
Adenosina/análogos & derivados , Antivirais/síntese química , Antivirais/farmacologia , Infecções por Arenaviridae/tratamento farmacológico , Arenaviridae/efeitos dos fármacos , Ebolavirus/efeitos dos fármacos , Doença pelo Vírus Ebola/tratamento farmacológico , Adenosina/química , Infecções por Arenaviridae/virologia , Doença pelo Vírus Ebola/virologia , Humanos , Estrutura Molecular
7.
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
8.
Antiviral Res ; 170: 104569, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31356830

RESUMO

Viral hemorrhagic fevers (VHFs) cause thousands of fatalities every year, but the treatment options for their management remain very limited. In particular, the development of therapeutic interventions is restricted by the lack of commercial viability of drugs targeting individual VHF agents. This makes approaches like drug repurposing and/or the identification of broad range therapies (i.e. those directed at host responses or common proviral factors) highly attractive. However, the identification of candidates for such antiviral repurposing or of host factors/pathways important for the virus life cycle is reliant on high-throughput screening (HTS). Recently, such screening work has been increasingly facilitated by the availability of reverse genetics-based approaches, including tools such as full-length clone (FLC) systems to generate reporter-expressing viruses or various life cycle modelling (LCM) systems, many of which have been developed and/or greatly improved during the last years. In particular, since LCM systems are capable of modelling specific steps in the life cycle, they are a valuable tool for both targeted screening (i.e. for inhibitors of a specific pathway) and mechanism of action studies. This review seeks to summarize the currently available reverse genetics systems for negative-sense VHF causing viruses (i.e. arenaviruses, bunyaviruses and filoviruses), and to highlight the recent advancements made in applying these systems for HTS to identify either antivirals or new virus-host interactions that might hold promise for the development of future treatments for the infections caused by these deadly but neglected virus groups.


Assuntos
Arenaviridae/genética , Bunyaviridae/genética , Filoviridae/genética , Febres Hemorrágicas Virais/virologia , Ensaios de Triagem em Larga Escala , Genética Reversa/métodos , Antivirais/isolamento & purificação , Arenaviridae/efeitos dos fármacos , Bunyaviridae/efeitos dos fármacos , Filoviridae/efeitos dos fármacos , Genoma Viral , Interações entre Hospedeiro e Microrganismos , Humanos
9.
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
10.
Expert Rev Anti Infect Ther ; 10(11): 1297-309, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23241187

RESUMO

Arenaviruses are a large group of emerging viruses including several causative agents of severe hemorrhagic fevers with high mortality in man. Considering the number of people affected and the currently limited therapeutic options, novel efficacious therapeutics against arenaviruses are urgently needed. Over the past decade, significant advances in knowledge about the basic virology of arenaviruses have been accompanied by the development of novel therapeutics targeting different steps of the arenaviral life cycle. High-throughput, small-molecule screens identified potent and broadly active inhibitors of arenavirus entry that were instrumental for the dissection of unique features of arenavirus fusion. Novel inhibitors of arenavirus replication have been successfully tested in animal models and hold promise for application in humans. Late in the arenavirus life cycle, the proteolytic processing of the arenavirus envelope glycoprotein precursor and cellular factors critically involved virion assembly and budding provide further promising 'druggable' targets for novel therapeutics to combat human arenavirus infection.


Assuntos
Antivirais/farmacologia , Antivirais/uso terapêutico , Infecções por Arenaviridae/tratamento farmacológico , Arenaviridae/efeitos dos fármacos , Arenaviridae/fisiologia , Descoberta de Drogas , Humanos , Proteínas do Envelope Viral , Proteínas Virais de Fusão/antagonistas & inibidores , Montagem de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
11.
Antiviral Res ; 91(2): 195-208, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21699921

RESUMO

Reverse-genetics systems are powerful tools enabling researchers to study the replication cycle of RNA viruses, including filoviruses and other hemorrhagic fever viruses, as well as to discover new antivirals. They include full-length clone systems as well as a number of life cycle modeling systems. Full-length clone systems allow for the generation of infectious, recombinant viruses, and thus are an important tool for studying the virus replication cycle in its entirety. In contrast, life cycle modeling systems such as minigenome and transcription and replication competent virus-like particle systems can be used to simulate and dissect parts of the virus life cycle outside of containment facilities. Minigenome systems are used to model viral genome replication and transcription, whereas transcription and replication competent virus-like particle systems also model morphogenesis and budding as well as infection of target cells. As such, these modeling systems have tremendous potential to further the discovery and screening of new antivirals targeting hemorrhagic fever viruses. This review provides an overview of currently established reverse genetics systems for hemorrhagic fever-causing negative-sense RNA viruses, with a particular emphasis on filoviruses, and the potential application of these systems for antiviral research.


Assuntos
Arenaviridae/genética , Bunyaviridae/genética , Filoviridae/genética , Genoma Viral , Antivirais/farmacologia , Arenaviridae/efeitos dos fármacos , Arenaviridae/fisiologia , Bunyaviridae/efeitos dos fármacos , Bunyaviridae/fisiologia , DNA Complementar/genética , DNA Complementar/metabolismo , Filoviridae/efeitos dos fármacos , Filoviridae/fisiologia , Genes Reporter , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Transcrição Gênica , Transfecção , Proteínas Virais/genética , Proteínas Virais/metabolismo , Internalização do Vírus , Liberação de Vírus , Replicação Viral
12.
Curr Mol Med ; 5(8): 735-51, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16375709

RESUMO

Arenaviruses merit significant attention both as tractable model systems to study acute and persistent viral infections, and as clinically important human pathogens. Evidence indicates that LCMV remains present in the USA and Europe and capable of causing significant morbidity in infected individuals, likely being a neglected human pathogen. Moreover, new arenaviruses are being discovered in the Americas on the average of one every three years, with some of them causing severe hemorrhagic fever. In addition, weaponized forms of these viruses pose a real threat as agents of bioterrorism. Therefore, it is important to develop effective vaccines and better antiviral drugs to combat the dual threats of naturally occurring and intentionally introduced Arenavirus infections. The development of arenavirus reverse genetic systems is allowing investigators to conduct a detailed molecular characterization of the viral cis-acting signals and trans-acting factors that control each of the steps of the Arenavirus life cycle, including RNA synthesis, packaging and budding. We will discuss how this new knowledge is facilitating the establishment of novel assays to identify and characterize compounds capable of interfering with specific steps of the virus life cycle. Likewise, the ability to generate predetermined specific mutations within the arenavirus genome, and analyze their phenotypic expression, would significantly contribute to the elucidation of arenavirus-host interactions, including the bases of their ability to persist, as well as to cause severe HF (hemorrhagic fever) disease in humans. These approaches could also lead to the development of novel potent and safe Arenavirus vaccines.


Assuntos
Anticorpos Antivirais/uso terapêutico , Antivirais/farmacologia , Infecções por Arenaviridae/tratamento farmacológico , Arenaviridae/efeitos dos fármacos , Vírus da Coriomeningite Linfocítica/efeitos dos fármacos , Vacinas Virais , Arenaviridae/genética , Arenaviridae/metabolismo , Infecções por Arenaviridae/epidemiologia , Infecções por Arenaviridae/prevenção & controle , Humanos , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/metabolismo
13.
J Med Chem ; 39(19): 3847-52, 1996 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-8809173

RESUMO

The syntheses and the antiviral activities of 2-halo derivatives of neplanocin A (1b,c), (6'R)-6'-C-methylneplanocin A (2b), and dehydroxymethylneplanocin A (3b,c) are described. SN2 reaction of the known cyclopentenyl units 12 and 13 with 2-haloadenines under basic conditions gave the protected carbocyclic nucleosides 14b,c and 15b,c, respectively. Starting from the cyclopentenone derivative 5, the optically active tosyloxycyclopentene derivative 11 was prepared, which was similarly condensed with 2-fluoroadenine to give the protected (6'R)-6'-C-methyl derivative 16b. Deprotection of these compounds afforded the target 2-halo derivatives of neplanocin A. Of these new compounds, 2-fluoroneplanocin A (1b) showed an antiviral potency and a spectrum that was comparable to that of neplanocin A (1a). It was particularly active against vaccinia virus, vesicular stomatitis virus, parainfluenza virus, reovirus, arenaviruses (Junin, Tacaribe), and human cytomegalovirus, i.e., those viruses that fall within the purview of the S-adenosyl-L-homocysteine hydrolase inhibitors.


Assuntos
Adenosina/análogos & derivados , Antivirais/síntese química , Antivirais/farmacologia , Adenosina/síntese química , Adenosina/química , Adenosina/farmacologia , Animais , Arenaviridae/efeitos dos fármacos , Chlorocebus aethiops , Citomegalovirus/efeitos dos fármacos , Estrutura Molecular , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Reoviridae/efeitos dos fármacos , Vaccinia virus/efeitos dos fármacos , Células Vero , Vírus da Estomatite Vesicular Indiana/efeitos dos fármacos
14.
Antiviral Res ; 18(2): 151-62, 1992 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-1365816

RESUMO

A novel nucleoside analog, 3'-fluoro-3'-deoxyadenosine (3'F3'dAdo), was evaluated for antiviral activity against several arthropod-borne and arenaviruses in Vero cell culture. The following 50% inhibitory concentrations (EC50) of virus plaque formation were obtained against the test viruses: Semliki Forest (10.3 microM) and Venezuelan equine encephalitis (5.3 microM) alphaviruses, lymphocytic choriomeningitis (7.7 microM) and Pichinde (greater than 32 microM) arenaviruses, Punta Toro (greater than 32 microM) and San Angelo (1.6 microM) bunyaviruses, banzi flavivirus (4.0 microM), and Colorado tick fever orbivirus (0.6 microM). By comparison, the broad-spectrum antiviral agent ribavirin was active against lymphocytic choriomeningitis (18 microM), Pichinde (24 microM), Punta Toro (114 microM), and San Angelo (99 microM) viruses, but was less active against the other 4 viruses (greater than 200 microM). Vero cell proliferation and thymidine and uridine incorporation into replicating Vero cells were inhibited by 50% with 3'F3'dAdo concentrations of 36, 45, and 32 microM, respectively. In virus yield reduction assays, increasing the multiplicity of infections of Semliki Forest and Venezuelan equine encephalitis viruses reduced the inhibitory activity of 3'F3'dAdo. Using the same assay, 3'F3'dAdo was found to enhance Punta Toro virus replication up to 5-fold relative to the untreated control. By adding the nucleoside transport inhibitor nitrobenzylthioinosine (100 microM) to the culture medium, antiviral activity against the two alphaviruses was eliminated, indicating that 3'F3'dAdo uses the nucleoside transport system for cell entry. When actinomycin D (5 microM) was used to greatly suppress cellular RNA synthesis in Semliki Forest virus-infected and uninfected cells, 3'F3'dAdo preferentially inhibited viral RNA synthesis. The results of these studies indicate 3'F3'dAdo is a selective inhibitor of most of the viruses tested and should be a promising candidate for in vivo evaluations.


Assuntos
Antivirais/farmacologia , Arbovírus/efeitos dos fármacos , Arenaviridae/efeitos dos fármacos , Desoxiadenosinas/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Chlorocebus aethiops , Depressão Química , Nucleosídeos/metabolismo , Phlebovirus/efeitos dos fármacos , RNA Viral/biossíntese , Vírus da Floresta de Semliki/efeitos dos fármacos , Vírus da Floresta de Semliki/metabolismo , Células Vero , Ensaio de Placa Viral
15.
Blood Cells ; 18(1): 7-25, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-1617194

RESUMO

In the presence of longwave ultraviolet light, psoralen derivatives photoreact with the nucleic acids within intact viruses and cells. This photoreaction can leave protein antigens and other surface components relatively unmodified, while eliminating the infectivity of a wide range of infectious agents. The kinetics of inactivation differ among RNA and DNA viruses photoreacted with different derivatives of psoralen. The inactivation kinetics are nonlinear as a result of photodegradation of psoralens and the unexplained biphasic inactivation of some viruses. In spite of these complexities, the photoreaction is capable of generating broad safety margins in the disinfection of microbial products under gentle, physiologic conditions. The psoralen photoreaction provides a potential method for inactivating both known and unknown viruses in active blood products. Psoralen-inactivated viruses have already proven useful as noninfectious antigens for use in immunoassays and as successful experimental vaccines.


Assuntos
DNA Viral/efeitos dos fármacos , Furocumarinas/farmacologia , RNA Viral/efeitos dos fármacos , Raios Ultravioleta , Vírus/efeitos dos fármacos , Antígenos Virais/efeitos dos fármacos , Antígenos Virais/efeitos da radiação , Arenaviridae/efeitos dos fármacos , Arenaviridae/fisiologia , Arenaviridae/efeitos da radiação , Dano ao DNA , DNA Viral/efeitos da radiação , Vírus da Encefalite Equina Venezuelana/efeitos dos fármacos , Vírus da Encefalite Equina Venezuelana/fisiologia , Vírus da Encefalite Equina Venezuelana/efeitos da radiação , Furocumarinas/efeitos da radiação , Cinética , Fotoquímica , RNA Viral/efeitos da radiação , Proteínas Virais/efeitos dos fármacos , Proteínas Virais/efeitos da radiação , Fenômenos Fisiológicos Virais , Replicação Viral/efeitos dos fármacos , Replicação Viral/efeitos da radiação , Vírus/imunologia , Vírus/efeitos da radiação
16.
Vopr Virusol ; 36(3): 231-5, 1991.
Artigo em Russo | MEDLINE | ID: mdl-1891878

RESUMO

The capacity of BHK-21 cell culture to produce Mopeia virus (Arenaviridae family) to form syncytium upon acidification of the culture medium to pH 5.5 and lower was demonstrated. The cell fusion requires their active virus production: a virus titre in the culture medium must be at least 10(5) PEU/ml. The inhibition of virus multiplication with ammonium chloride as well as treatment of the cells before the medium acidification with immune serum reduced syncytium formation markedly. No cell fusion was observed upon acidification of the medium immediately after virus adsorption to cells. Thus, the observed cell fusion under the influence of the virus is an "internal fusion" and confirms our previous data on the endocytosis mode of arenavirus penetration into the cell.


Assuntos
Compostos de Alumínio , Arenaviridae/patogenicidade , Adsorção , Alumínio/farmacologia , Cloreto de Alumínio , Animais , Arenaviridae/efeitos dos fármacos , Arenaviridae/imunologia , Fusão Celular/efeitos dos fármacos , Linhagem Celular , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/microbiologia , Cloretos/farmacologia , Cricetinae , Células Gigantes/citologia , Células Gigantes/efeitos dos fármacos , Células Gigantes/microbiologia , Concentração de Íons de Hidrogênio , Soros Imunes/farmacologia , Cultura de Vírus
17.
Antiviral Res ; 14(4-5): 287-99, 1990.
Artigo em Inglês | MEDLINE | ID: mdl-2088208

RESUMO

Several compounds, belonging to different classes of nucleoside analogues and sulfated polysaccharides, were evaluated for their inhibitory effects on the replication of the arenaviruses Junin and Tacaribe in VERO cells. S-Adenosylhomocysteine (AdoHcy) hydrolase inhibitors [i.e. adenosine dialdehyde, carbocyclic 3-deazaadenosine (C-c3 Ado), neplanocin A, 3-deazaneplanocin A, 9-(2,3-dihydroxypropyl)adenine [(S)-DHPA], (RS)-3-adenin-9-yl-hydroxypropanoic acid isobutyl ester [(RS)-AHPA], the 2',3'-dihydroxycyclopentenyl derivatives of adenine (DHCA) and 3-deazaadenine (DHCDA)] inhibited arenavirus replication within the concentration range of 1-10 micrograms/ml, while not being toxic for cell morphology or cellular DNA synthesis at a concentration of 100-400 micrograms/ml. Based on the ratio of the concentrations required to inhibit cell proliferation and virus replication, only (S)-DHPA, DHCA, C-c3 Ado and adenosine dialdehyde could be considered as truly selective inhibitors. Tubercidin, cyclopentenyl cytosine, pyrazofurin and ribavirin also inhibited viral cytopathogenicity at concentrations that were well below the cytotoxic threshold. Carbodine (cyclopentyl cytosine) also proved to be a potent inhibitor of arenavirus replication, but it was not as selective as cyclopentenyl cytosine. Very potent and selective inhibitors were the sulfated polysaccharides dextran sulfate, lambda-carrageenan, fucoidan, heparin and pentosan polysulfate: they inhibited virus replication at a concentration of 0.1-2.8 micrograms/ml, whereas the compounds were not inhibitory to cell growth even at a concentration of 200 micrograms/ml.


Assuntos
Antivirais/farmacologia , Arenaviridae/efeitos dos fármacos , Nucleosídeos/farmacologia , Reagentes de Sulfidrila/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Arenaviridae/crescimento & desenvolvimento , Efeito Citopatogênico Viral , DNA Viral/biossíntese , Células Vero/microbiologia
19.
Vopr Virusol ; 35(2): 146-50, 1990.
Artigo em Russo | MEDLINE | ID: mdl-2389568

RESUMO

Lysosomotropic agents (NH4Cl, amantadine, chloroquine, monensin) which prevent acidification of intracellular vacuoles, when introduced into the culture medium before or during inoculation of cells (BHK-21, Vero) with arenaviruses inhibit reproduction of these viruses completely or significantly. Mozambique virus proved to be 10 times more sensitive to the effect of lysosomotropic agents than Pichinde and Lassa viruses. Thus, arenaviruses have a pH-dependent stage at the beginning of the reproduction cycle which is indirectly indicative of their penetration into cells by receptor endocytosis.


Assuntos
Arenaviridae/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Adsorção , Amantadina/farmacologia , Cloreto de Amônio/farmacologia , Animais , Arenaviridae/patogenicidade , Arenaviridae/fisiologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/microbiologia , Cloroquina/farmacologia , Concentração de Íons de Hidrogênio , Vírus Lassa/efeitos dos fármacos , Vírus Lassa/patogenicidade , Vírus Lassa/fisiologia , Monensin/farmacologia , Fatores de Tempo , Células Vero/efeitos dos fármacos , Células Vero/microbiologia , Replicação Viral/efeitos dos fármacos
20.
J Med Virol ; 29(2): 146-51, 1989 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2557385

RESUMO

Administration of cyclophosphamide (CY) to guinea pigs infected with a lethal strain of Junin virus (JV) delayed the time of death, with survival of a small number of animals. Virological studies showed a temporary decrease of virus concentration in blood and viscera shortly after the CY injection. In the pathological study no differences were found in the organic lesions present in CY-treated and nontreated animals, with the exception of the pulmonary alterations. In CY-treated guinea pigs the lungs appeared almost normal, but in the control, nontreated animals severe alterations with the pattern of the "respiratory distress syndrome of the adult" were consistently present. In in vitro experiments, incorporation of serum collected from guinea pigs injected 30 minutes before exsanguination with CY to cell cultures, infected with JV, prevented virus replication. On the basis of these results it is suggested that the delay of time of death and eventual survival of CY-treated guinea pigs after JV infection depends on a direct antiviral effect of the drug rather than on its known immunosuppressive action. In addition, the absence of pulmonary alterations in CY-treated animals was tentatively considered to be dependent on the marked polymorphonuclear leukocyte depletion induced by the drug.


Assuntos
Arenaviridae/efeitos dos fármacos , Arenavirus do Novo Mundo/efeitos dos fármacos , Ciclofosfamida/uso terapêutico , Febre Hemorrágica Americana/tratamento farmacológico , Animais , Arenavirus do Novo Mundo/crescimento & desenvolvimento , Sobrevivência Celular , Células Cultivadas , Relação Dose-Resposta a Droga , Cobaias , Febre Hemorrágica Americana/patologia , Masculino , Especificidade de Órgãos , Ensaio de Placa Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA