Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Negl Trop Dis ; 7(9): e2430, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24069485

RESUMO

For enveloped viruses, fusion of the viral envelope with a cellular membrane is critical for a productive infection to occur. This fusion process is mediated by at least three classes of fusion proteins (Class I, II, and III) based on the protein sequence and structure. For Rift Valley fever virus (RVFV), the glycoprotein Gc (Class II fusion protein) mediates this fusion event following entry into the endocytic pathway, allowing the viral genome access to the cell cytoplasm. Here, we show that peptides analogous to the RVFV Gc stem region inhibited RVFV infectivity in cell culture by inhibiting the fusion process. Further, we show that infectivity can be inhibited for diverse, unrelated RNA viruses that have Class I (Ebola virus), Class II (Andes virus), or Class III (vesicular stomatitis virus) fusion proteins using this single peptide. Our findings are consistent with an inhibition mechanism similar to that proposed for stem peptide fusion inhibitors of dengue virus in which the RVFV inhibitory peptide first binds to both the virion and cell membranes, allowing it to traffic with the virus into the endocytic pathway. Upon acidification and rearrangement of Gc, the peptide is then able to specifically bind to Gc and prevent fusion of the viral and endocytic membranes, thus inhibiting viral infection. These results could provide novel insights into conserved features among the three classes of viral fusion proteins and offer direction for the future development of broadly active fusion inhibitors.


Assuntos
Antivirais/metabolismo , Bunyaviridae/fisiologia , Mononegavirais/fisiologia , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus , Animais , Bunyaviridae/efeitos dos fármacos , Chlorocebus aethiops , Ebolavirus/efeitos dos fármacos , Ebolavirus/fisiologia , Mononegavirais/efeitos dos fármacos , Células Vero
2.
Vaccines (Basel) ; 1(3): 262-77, 2013 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-26344112

RESUMO

Lassa virus (LASV) causes a severe, often fatal, hemorrhagic fever endemic to West Africa. Presently, there are no FDA-licensed medical countermeasures for this disease. In a pilot study, we constructed a DNA vaccine (pLASV-GPC) that expressed the LASV glycoprotein precursor gene (GPC). This plasmid was used to vaccinate guinea pigs (GPs) using intramuscular electroporation as the delivery platform. Vaccinated GPs were protected from lethal infection (5/6) with LASV compared to the controls. However, vaccinated GPs experienced transient viremia after challenge, although lower than the mock-vaccinated controls. In a follow-on study, we developed a new device that allowed for both the vaccine and electroporation pulse to be delivered to the dermis. We also codon-optimized the GPC sequence of the vaccine to enhance expression in GPs. Together, these innovations resulted in enhanced efficacy of the vaccine. Unlike the pilot study where neutralizing titers were not detected until after virus challenge, modest neutralizing titers were detected in guinea pigs before challenge, with escalating titers detected after challenge. The vaccinated GPs were never ill and were not viremic at any timepoint. The combination of the codon-optimized vaccine and dermal electroporation delivery is a worthy candidate for further development.

3.
Antiviral Res ; 90(1): 70-9, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21371508

RESUMO

Lassa virus (LASV), a member of the Arenaviridae family, causes a viral hemorrhagic fever endemic to West Africa, where as many as 300,000 infections occur per year. Presently, there are no FDA-approved LASV-specific vaccines or antiviral agents, although the antiviral drug ribavirin has shown some efficacy. A recently identified small-molecule inhibitor of arenavirus entry, ST-193, exhibits submicromolar antiviral activity in vitro. To determine the antiviral utility of ST-193 in vivo, we tested the efficacy of this compound in the LASV guinea pig model. Four groups of strain 13 guinea pigs were administered 25 or 80 mg/kg ST-193, 25 mg/kg of ribavirin, or the vehicle by the intraperitoneal (i.p.) route before infection with a lethal dose of LASV, strain Josiah, and continuing once daily for 14 days. Control animals exhibited severe disease, becoming moribund between days 10 and 15 postinfection. ST-193-treated animals exhibited fewer signs of disease and enhanced survival when compared to the ribavirin or vehicle groups. Body temperatures in all groups were elevated by day 9, but returned to normal by day 19 postinfection in the majority of ST-193-treated animals. ST-193 treatment mediated a 2-3-log reduction in viremia relative to vehicle-treated controls. The overall survival rate for the ST-193-treated guinea pigs was 62.5% (10/16) compared with 0% in the ribavirin (0/8) and vehicle (0/7) groups. These data suggest that ST-193 may serve as an improved candidate for the treatment of Lassa fever.


Assuntos
Antivirais/administração & dosagem , Febre Lassa/tratamento farmacológico , Animais , Temperatura Corporal , Modelos Animais de Doenças , Feminino , Cobaias , Injeções Intraperitoneais , Febre Lassa/mortalidade , Febre Lassa/patologia , Análise de Sobrevida , Viremia/prevenção & controle
4.
Virol J ; 5: 161, 2008 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-19105844

RESUMO

BACKGROUND: Sera from convalescent Lassa fever patients often contains antibodies to Lassa virus (LASV) glycoprotein 1 (GP1), and glycoprotein 2 (GP2); Immunization of non-human primates with viral vectors expressing the arenaviral glycoprotein complex (GPC) confers full protective immunity against a lethal challenge with LASV. Thus, the development of native or quasi native recombinant LASV GP1 and GP2 as soluble, uncoupled proteins will improve current diagnostics, treatment, and prevention of Lassa fever. To this end, mammalian expression systems were engineered for production and purification of secreted forms of soluble LASV GP1 and GP2 proteins. RESULTS: Determinants for mammalian cell expression of secreted uncoupled Lassa virus (LASV) glycoprotein 1 (GP1) and glycoprotein 2 (GP2) were established. Soluble GP1 was generated using either the native glycoprotein precursor (GPC) signal peptide (SP) or human IgG signal sequences (s.s.). GP2 was secreted from cells only when (1) the transmembrane (TM) domain was deleted, the intracellular domain (IC) was fused to the ectodomain, and the gene was co-expressed with a complete GP1 gene in cis; (2) the TM and IC domains were deleted and GP1 was co-expressed in cis; (3) expression of GP1 was driven by the native GPC SP. These data implicate GP1 as a chaperone for processing and shuttling GP2 to the cell surface. The soluble forms of GP1 and GP2 generated through these studies were secreted as homogeneously glycosylated proteins that contained high mannose glycans. Furthermore, observation of GP1 ectodomain shedding from cells expressing wild type LASV GPC represents a novel aspect of arenaviral glycoprotein expression. CONCLUSION: These results implicate GP1 as a chaperone for the correct processing and shuttling of GP2 to the cell surface, and suggest that native GPC SP plays a role in this process. In the absence of GP1 and GPC SP the GP2 protein may be processed by an alternate pathway that produces heterogeneously glycosylated protein, or the polypeptide may not fully mature in the secretory cascade in mammalian cells. The expression constructs developed in these studies resulted in the generation and purification of soluble, uncoupled GP1 and GP2 proteins from mammalian cells with quasi-native properties. The observation of GP1 ectodomain shedding from cells expressing wild type LASV GPC establishes new correlates of disease progression and highlights potential opportunities for development of diagnostics targeting the early stages of Lassa fever.


Assuntos
Regulação Viral da Expressão Gênica , Vírus Lassa/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Glicosilação , Humanos , Vírus Lassa/genética , Células Vero , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/isolamento & purificação
5.
Virol J ; 5: 74, 2008 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-18538016

RESUMO

BACKGROUND: There is a significant requirement for the development and acquisition of reagents that will facilitate effective diagnosis, treatment, and prevention of Lassa fever. In this regard, recombinant Lassa virus (LASV) proteins may serve as valuable tools in diverse antiviral applications. Bacterial-based systems were engineered for expression and purification of recombinant LASV nucleoprotein (NP), glycoprotein 1 (GP1), and glycoprotein 2 (GP2). RESULTS: Full-length NP and the ectodomains of GP1 and GP2 were generated as maltose-binding protein (MBP) fusions in the Rosetta strains of Escherichia coli (E. coli) using pMAL-c2x vectors. Average fusion protein yields per liter of culture for MBP-NP, MBP-GP1, and MBP-GP2 were 10 mg, 9 mg, and 9 mg, respectively. Each protein was captured from cell lysates using amylose resin, cleaved with Factor Xa, and purified using size-exclusion chromatography (SEC). Fermentation cultures resulted in average yields per liter of 1.6 mg, 1.5 mg, and 0.7 mg of purified NP, GP1 and GP2, respectively. LASV-specific antibodies in human convalescent sera specifically detected each of the purified recombinant LASV proteins, highlighting their utility in diagnostic applications. In addition, mouse hyperimmune ascitic fluids (MHAF) against a panel of Old and New World arenaviruses demonstrated selective cross reactivity with LASV proteins in Western blot and enzyme-linked immunosorbent assay (ELISA). CONCLUSION: These results demonstrate the potential for developing broadly reactive immunological assays that employ all three arenaviral proteins individually and in combination.


Assuntos
Antígenos Virais/biossíntese , Antígenos Virais/isolamento & purificação , Vírus Lassa/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Virais/biossíntese , Proteínas Virais/isolamento & purificação , Animais , Anticorpos Antibacterianos/sangue , Antígenos Virais/genética , Arenavirus/imunologia , Proteínas do Capsídeo/biossíntese , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/isolamento & purificação , Cromatografia de Afinidade , Reações Cruzadas , Escherichia coli/genética , Humanos , Camundongos , Proteínas Recombinantes de Fusão/genética , Proteínas do Envelope Viral/biossíntese , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/isolamento & purificação , Proteínas Virais/genética
6.
Antiviral Res ; 78(1): 103-15, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18241935

RESUMO

Unlike many viral hemorrhagic fevers (VHFs), Lassa fever (LF) is not a rare disease that emerges only as sporadic cases or in outbreak form. Although surveillance is inadequate to determine the true incidence, up to 300,000 infections and 5000 deaths from LF are estimated to occur yearly. The highest incidence is in the "Mano River Union (MRU) countries" of Sierra Leone, Liberia, and Guinea. Although civil unrest in this region over the past two decades has impeded capacity building and research, new-found peace in recent years presents new opportunities. In 2004, the Mano River Union Lassa Fever Network (MRU LFN) was established to assist MRU countries in the development of national and regional surveillance, diagnosis, treatment, control, and prevention of LF. Here, we review the present literature on treatment and pathogenesis of LF and outline priorities for future research in the field made possible by the improved research capacity of the MRU LFN.


Assuntos
Antivirais/uso terapêutico , Febre Lassa/tratamento farmacológico , Febre Lassa/fisiopatologia , Vírus Lassa/patogenicidade , Ribavirina/uso terapêutico , Adolescente , Adulto , África Ocidental/epidemiologia , Pré-Escolar , Feminino , Humanos , Incidência , Recém-Nascido , Febre Lassa/epidemiologia , Febre Lassa/virologia , Vírus Lassa/efeitos dos fármacos , Masculino , Gravidez , Pesquisa/tendências
7.
J Virol ; 81(16): 8634-47, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17537852

RESUMO

In contrast to most negative-stranded RNA viruses, hantaviruses and other viruses in the family Bunyaviridae mature intracellularly, deriving the virion envelope from the endoplasmic reticulum (ER) or Golgi compartment. While it is generally accepted that Old World hantaviruses assemble and bud into the Golgi compartment, some studies with New World hantaviruses have raised the possibility of maturation at the plasma membrane as well. Overall, the steps leading to virion assembly remain largely undetermined for hantaviruses. Because hantaviruses do not have matrix proteins, the nucleocapsid protein (N) has been proposed to play a key role in assembly. Herein, we examine the intracellular trafficking and morphogenesis of the prototype Old World hantavirus, Hantaan virus (HTNV). Using confocal microscopy, we show that N colocalized with the ER-Golgi intermediate compartment (ERGIC) in HTNV-infected Vero E6 cells, not with the ER, Golgi compartment, or early endosomes. Brefeldin A, which effectively disperses the ER, the ERGIC, and Golgi membranes, redistributed N with the ERGIC, implicating membrane association; however, subcellular fractionation experiments showed the majority of N in particulate fractions. Confocal microscopy revealed that N was juxtaposed to and distributed along microtubules and, over time, became surrounded by vimentin cages. To probe cytoskeletal association further, we probed trafficking of N in cells treated with nocodazole and cytochalasin D, which depolymerize microtubules and actin, respectively. We show that nocodazole, but not cytochalasin D, affected the distribution of N and reduced levels of intracellular viral RNA. These results suggested the involvement of microtubules in trafficking of N, whose movement could occur via molecular motors such as dynein. Overexpression of dynamitin, which is associated with dynein-mediated transport, creates a dominant-negative phenotype blocking transport on microtubules. Overexpression of dynamitin reduced N accumulation in the perinuclear region, which further supports microtubule components in N trafficking. The combined results of these experiments support targeting of N to the ERGIC prior to its movement to the Golgi compartment and the requirement of an intact ERGIC for viral replication and, thus, the possibility of virus factories in this region.


Assuntos
Proteínas do Capsídeo/metabolismo , Dineínas/metabolismo , Retículo Endoplasmático/virologia , Complexo de Golgi/virologia , Vírus Hantaan/crescimento & desenvolvimento , Proteínas do Core Viral/metabolismo , Animais , Proteínas do Capsídeo/análise , Membrana Celular/química , Membrana Celular/metabolismo , Chlorocebus aethiops , Citocalasina D/farmacologia , Complexo Dinactina , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/ultraestrutura , Complexo de Golgi/metabolismo , Complexo de Golgi/ultraestrutura , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Nocodazol/farmacologia , Inibidores da Síntese de Ácido Nucleico/farmacologia , Transporte Proteico/efeitos dos fármacos , Coelhos , Células Vero , Vimentina/metabolismo , Proteínas do Core Viral/análise
8.
Antiviral Res ; 69(2): 86-97, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16343651

RESUMO

Category A arenaviruses as defined by the National Institute of Allergy and Infectious Diseases (NIAID) are human pathogens that could be weaponized by bioterrorists. Many of these deadly viruses require biosafety level-4 (BSL-4) containment for all laboratory work, which limits traditional laboratory high-throughput screening (HTS) for identification of small molecule inhibitors. For those reasons, a related BSL-2 New World arenavirus, Tacaribe virus, 67-78% identical to Junín virus at the amino acid level, was used in a HTS campaign where approximately 400,000 small molecule compounds were screened in a Tacaribe virus-induced cytopathic effect (CPE) assay. Compounds identified in this screen showed antiviral activity and specificity against not only Tacaribe virus, but also the Category A New World arenaviruses (Junín, Machupo, and Guanarito). Drug resistant variants were isolated, suggesting that these compounds act through inhibition of a viral protein, the viral glycoprotein (GP2), and not through cellular toxicity mechanisms. A lead compound, ST-294, has been chosen for drug development. This potent and selective compound, with good bioavailability, demonstrated protective anti-viral efficacy in a Tacaribe mouse challenge model. This series of compounds represent a new class of inhibitors that may warrant further development for potential inclusion in a strategic stockpile.


Assuntos
Antivirais/química , Arenavirus do Novo Mundo/efeitos dos fármacos , Chumbo/química , Proteínas Virais/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Antivirais/farmacologia , Infecções por Arenaviridae/tratamento farmacológico , Infecções por Arenaviridae/virologia , Chlorocebus aethiops , Efeito Citopatogênico Viral , Febres Hemorrágicas Virais/tratamento farmacológico , Febres Hemorrágicas Virais/virologia , Humanos , Chumbo/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Ratos , Ratos Sprague-Dawley , Sulfonamidas/química , Sulfonamidas/farmacologia , Ureia/análogos & derivados , Ureia/química , Ureia/farmacologia , Células Vero , Proteínas Virais/metabolismo
9.
Vaccine ; 24(21): 4657-66, 2006 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-16174542

RESUMO

DNA vaccines for Rift Valley fever virus (RVFV), Crimean Congo hemorrhagic fever virus (CCHFV), tick-borne encephalitis virus (TBEV), and Hantaan virus (HTNV), were tested in mice alone or in various combinations. The bunyavirus vaccines (RVFV, CCHFV, and HTNV) expressed Gn and Gc genes, and the flavivirus vaccine (TBEV) expressed the preM and E genes. All vaccines were delivered by gene gun. The TBEV DNA vaccine and the RVFV DNA vaccine elicited similar levels of antibodies and protected mice from challenge when delivered alone or in combination with other DNAs. Although in general, the HTNV and CCHFV DNA vaccines were not very immunogenic in mice, there were no major differences in performance when given alone or in combination with the other vaccines.


Assuntos
Vírus da Encefalite Transmitidos por Carrapatos/imunologia , Vírus da Febre Hemorrágica da Crimeia-Congo/imunologia , Vírus da Febre do Vale do Rift/imunologia , Vacinas Combinadas/imunologia , Vacinas de DNA/imunologia , Animais , Sequência de Bases , Células COS , Chlorocebus aethiops , Primers do DNA , Vírus da Encefalite Transmitidos por Carrapatos/crescimento & desenvolvimento , Vírus da Febre Hemorrágica da Crimeia-Congo/crescimento & desenvolvimento , Camundongos , Testes de Neutralização , Vírus da Febre do Vale do Rift/crescimento & desenvolvimento , Ensaio de Placa Viral
10.
PLoS Med ; 2(6): e183, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15971954

RESUMO

BACKGROUND: Recent importation of Lassa fever into Germany, the Netherlands, the United Kingdom, and the United States by travelers on commercial airlines from Africa underscores the public health challenge of emerging viruses. Currently, there are no licensed vaccines for Lassa fever, and no experimental vaccine has completely protected nonhuman primates against a lethal challenge. METHODS AND FINDINGS: We developed a replication-competent vaccine against Lassa virus based on attenuated recombinant vesicular stomatitis virus vectors expressing the Lassa viral glycoprotein. A single intramuscular vaccination of the Lassa vaccine elicited a protective immune response in nonhuman primates against a lethal Lassa virus challenge. Vaccine shedding was not detected in the monkeys, and none of the animals developed fever or other symptoms of illness associated with vaccination. The Lassa vaccine induced strong humoral and cellular immune responses in the four vaccinated and challenged monkeys. Despite a transient Lassa viremia in vaccinated animals 7 d after challenge, the vaccinated animals showed no evidence of clinical disease. In contrast, the two control animals developed severe symptoms including rashes, facial edema, and elevated liver enzymes, and ultimately succumbed to the Lassa infection. CONCLUSION: Our data suggest that the Lassa vaccine candidate based on recombinant vesicular stomatitis virus is safe and highly efficacious in a relevant animal model that faithfully reproduces human disease.


Assuntos
Febre Lassa/prevenção & controle , Vírus Lassa/imunologia , Vacinas Virais , Animais , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Febre Lassa/imunologia , Macaca fascicularis , Vacinação , Vacinas Sintéticas , Vacinas Virais/imunologia , Viremia/prevenção & controle
12.
Hybrid Hybridomics ; 22(3): 135-45, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12954098

RESUMO

Phage display technology allows for the production and rapid selection of antigen-specific, Fab antibody fragments. For purposes of immune therapy, though, complete antibodies that retain the Fc domain are often required. In this regard, we designed cassette vectors for converting human Fab fragments selected from combinatorial phage display libraries into full-length IgG(1) monoclonal antibodies (MAbs). Two expression vectors, pIEI-Light and pIEI-Heavy, were engineered to contain respective light- and heavy-chain human signal sequences downstream of the baculovirus immediate early gene promoter, IEI. Vector pIEI-Heavy also contains the coding region for each of the human IgG(1) constant domains. To generate complete antibody genes, the cassette vectors possess convenient restriction enzyme sites for rapid in-frame cloning of coding regions for full-length light chains in pIEI-Light and for the heavy-chain variable domains in pIEI-Heavy of Fab fragments. Using these constructs and a method that allows for stable transformation of insect cells, complete light- and heavy-chain genes can be inserted into the insect cell genome and subsequently expressed under the control of the baculovirus IEI promoter. This cassette vector system was used to generate stably transformed insect cells that continuously secreted functional full-length, IgG(1) MAbs. The expressed antibodies exhibited light and heavy chains of the appropriate molecular sizes and retained the ability to bind antigen. We conclude that our cassette vectors could serve as valuable tools for generating human IgG(1) antibodies.


Assuntos
Anticorpos Monoclonais/imunologia , Vetores Genéticos , Fragmentos Fab das Imunoglobulinas/genética , Imunoglobulina G/imunologia , Sequência de Aminoácidos , Animais , Baculoviridae/genética , Sequência de Bases , Linhagem Celular Transformada , Chlorocebus aethiops , DNA/química , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Genes de Imunoglobulinas/genética , Imunoglobulina G/química , Insetos , Modelos Genéticos , Dados de Sequência Molecular , Biblioteca de Peptídeos , Testes de Precipitina , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...