Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Vaccines (Basel) ; 8(3)2020 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-32664486

RESUMO

Hantaan virus (HTNV) and Puumala virus (PUUV) are pathogenic hantaviruses found in Asia and Europe, respectively. DNA vaccines targeting the envelope glycoproteins of these viruses have been constructed and found to elicit neutralizing antibodies when delivered to humans by various technologies including intramuscular electroporation. Here, we report findings from a Phase 2a clinical trial of a combined HTNV/PUUV DNA vaccine delivered at varying doses and administration schedules using the Ichor Medical Systems TriGrid intramuscular electroporation delivery technology. The study was designed to characterize the effects of DNA vaccine dose and number of administrations on the frequency and magnitude of immunological response. Subjects (n = 120) were divided into four cohorts. Cohorts 1 and 2 received a dose of 2 mg of DNA (1 mg per plasmid), and cohorts 3 and 4 received a dose of 1 mg of DNA (0.5 mg per plasmid) each vaccination. Each of the four cohorts received a series of four administrations (days 0, 28, 56 and 168). For cohorts 1 and 3, the DNA vaccine candidate was delivered at each of the four administrations. For cohorts 2 and 4, in order to maintain blinding, subjects received the DNA vaccine on days 0, 56 and 168, but on day 28 received only the phosphate buffered saline vehicle rather the DNA vaccine. Sera were collected on days 0, 28, 56, 84, 140, 168, 196, 252 and 365 and evaluated for the presence of neutralizing antibodies by PUUV and HTNV pseudovirion neutralization assays (PsVNAs). Day 84 was also evaluated by a plaque reduction neutralization test (PRNT). Overall the PsVNA50 geometric mean titers (GMTs) and seropositivity rates among cohorts were similar. Cohort 3 exhibited the highest frequency of subjects that became seropositive to both PUUV and HTNV after vaccination, the highest peak GMT against both viruses, and the highest median titers against both viruses.

2.
J Virol ; 93(8)2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30700611

RESUMO

Antiviral therapies that impede virus entry are attractive because they act on the first phase of the infectious cycle. Drugs that target pathways common to multiple viruses are particularly desirable when laboratory-based viral identification may be challenging, e.g., in an outbreak setting. We are interested in identifying drugs that block both Ebola virus (EBOV) and Lassa virus (LASV), two unrelated but highly pathogenic hemorrhagic fever viruses that have caused outbreaks in similar regions in Africa and share features of virus entry: use of cell surface attachment factors, macropinocytosis, endosomal receptors, and low pH to trigger fusion in late endosomes. Toward this goal, we directly compared the potency of eight drugs known to block EBOV entry with their potency as inhibitors of LASV entry. Five drugs (amodiaquine, apilimod, arbidol, niclosamide, and zoniporide) showed roughly equivalent degrees of inhibition of LASV and EBOV glycoprotein (GP)-bearing pseudoviruses; three (clomiphene, sertraline, and toremifene) were more potent against EBOV. We then focused on arbidol, which is licensed abroad as an anti-influenza drug and exhibits activity against a diverse array of clinically relevant viruses. We found that arbidol inhibits infection by authentic LASV, inhibits LASV GP-mediated cell-cell fusion and virus-cell fusion, and, reminiscent of its activity on influenza virus hemagglutinin, stabilizes LASV GP to low-pH exposure. Our findings suggest that arbidol inhibits LASV fusion, which may partly involve blocking conformational changes in LASV GP. We discuss our findings in terms of the potential to develop a drug cocktail that could inhibit both LASV and EBOV.IMPORTANCE Lassa and Ebola viruses continue to cause severe outbreaks in humans, yet there are only limited therapeutic options to treat the deadly hemorrhagic fever diseases they cause. Because of overlapping geographic occurrences and similarities in mode of entry into cells, we seek a practical drug or drug cocktail that could be used to treat infections by both viruses. Toward this goal, we directly compared eight drugs, approved or in clinical testing, for the ability to block entry mediated by the glycoproteins of both viruses. We identified five drugs with approximately equal potencies against both. Among these, we investigated the modes of action of arbidol, a drug licensed abroad to treat influenza infections. We found, as shown for influenza virus, that arbidol blocks fusion mediated by the Lassa virus glycoprotein. Our findings encourage the development of a combination of approved drugs to treat both Lassa and Ebola virus diseases.


Assuntos
Antivirais/farmacologia , Ebolavirus/metabolismo , Doença pelo Vírus Ebola/tratamento farmacológico , Indóis/farmacologia , Febre Lassa/tratamento farmacológico , Vírus Lassa/metabolismo , Animais , Células COS , Chlorocebus aethiops , Cricetinae , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Doença pelo Vírus Ebola/metabolismo , Doença pelo Vírus Ebola/patologia , Humanos , Febre Lassa/metabolismo , Febre Lassa/patologia , Células Vero , Internalização do Vírus/efeitos dos fármacos
3.
Clin Vaccine Immunol ; 20(2): 218-26, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23239797

RESUMO

Puumala virus (PUUV) is a causative agent of hemorrhagic fever with renal syndrome (HFRS). Although PUUV-associated HFRS does not result in high case-fatality rates, the social and economic impact is considerable. There is no licensed vaccine or specific therapeutic to prevent or treat HFRS. Here we report the synthesis of a codon-optimized, full-length M segment open reading frame and its cloning into a DNA vaccine vector to produce the plasmid pWRG/PUU-M(s2). pWRG/PUU-M(s2) delivered by gene gun produced high-titer neutralizing antibodies in hamsters and nonhuman primates. Vaccination with pWRG/PUU-M(s2) protected hamsters against infection with PUUV but not against infection by related HFRS-associated hantaviruses. Unexpectedly, vaccination protected hamsters in a lethal disease model of Andes virus (ANDV) in the absence of ANDV cross-neutralizing antibodies. This is the first evidence that an experimental DNA vaccine for HFRS can provide protection in a hantavirus lethal disease model.


Assuntos
Infecções por Hantavirus/imunologia , Febre Hemorrágica com Síndrome Renal/imunologia , Virus Puumala/imunologia , Vacinas de DNA/imunologia , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Células COS , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Reações Cruzadas , DNA Viral/imunologia , Orthohantavírus/imunologia , Febre Hemorrágica com Síndrome Renal/prevenção & controle , Febre Hemorrágica com Síndrome Renal/virologia , Macaca mulatta/imunologia , Testes de Neutralização , Vacinação , Vacinas de DNA/administração & dosagem , Células Vero , Ensaio de Placa Viral , Vacinas Virais/administração & dosagem
4.
Vaccine ; 29(39): 6728-35, 2011 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-21219978

RESUMO

We have developed a rapid, reliable, and sensitive quantitative flow cytometric assay to measure the in vitro potency and stability of DNA vaccines to be delivered either by particle-mediated epidermal delivery (PMED) or by electroporation. The method involves transfecting cells with test DNA and comparing the measured antigen expression to that generated with expression from known quantities of reference material DNA. The assay was adapted for performance under Good Laboratory Practice (GLP) guidelines and was successfully utilized to perform potency testing in support of a Phase I study for two hantavirus DNA vaccines delivered by gene gun. The results from the potency assays conducted over a 24-month period using this method proved to be highly reproducible with high signal-to-noise ratios. The assay was also adapted to assess the in vitro potency and stability of a DNA vaccine for Venezuelan equine encephalitis virus that will be delivered by electroporation. Our results indicate that this assay can be readily applied to support potency and stability testing of numerous DNA vaccines delivered by various methods, including multiagent vaccines.


Assuntos
Citometria de Fluxo/métodos , Orthohantavírus/imunologia , Vacinas de DNA/imunologia , Animais , Antígenos Virais/imunologia , Biolística , Células COS , Chlorocebus aethiops , Sistemas de Liberação de Medicamentos/métodos , Estabilidade de Medicamentos , Eletroforese em Gel de Ágar , Eletroporação , Vírus da Encefalite Equina Venezuelana/genética , Vírus da Encefalite Equina Venezuelana/imunologia , Citometria de Fluxo/instrumentação , Orthohantavírus/genética , Humanos , Plasmídeos/genética , Plasmídeos/metabolismo , Padrões de Referência , Reprodutibilidade dos Testes , Transfecção , Vacinas de DNA/administração & dosagem , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia
5.
Virology ; 326(1): 130-9, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15262501

RESUMO

Sin Nombre virus (SNV) and Andes virus (ANDV), members of the genus Hantavirus, in the family Bunyaviridae, are causative agents of hantavirus pulmonary syndrome (HPS) in North and South America, respectively. Although ANDV causes a lethal HPS-like disease in hamsters, SNV, and all other HPS-associated hantaviruses that have been tested, cause asymptomatic infections of laboratory animals, including hamsters. In an effort to understand the pathogenicity of ANDV in the hamster model, we generated ANDV/SNV reassortant viruses. Plaque isolation of viruses from cell cultures infected with both parental viruses yielded only one type of stable reassortant virus: large (L) and small (S) segments of SNV and M segment of ANDV. This virus, designated SAS reassortant virus, had in vitro growth and plaque morphology characteristics similar to those of ANDV. When injected into hamsters, the SAS reassortant virus was highly infectious and elicited high-titer, ANDV-specific neutralizing antibodies; however, the virus did not cause HPS and was not lethal. These data indicate that the ANDV M genome segment is not sufficient to confer the lethal HPS phenotype associated with ANDV.


Assuntos
Genoma Viral , Infecções por Hantavirus/virologia , Orthohantavírus/fisiologia , Vírus Reordenados/fisiologia , Animais , Chlorocebus aethiops , Cricetinae , Modelos Animais de Doenças , Feminino , Orthohantavírus/genética , Orthohantavírus/patogenicidade , Mesocricetus , Vírus Reordenados/patogenicidade , Vírus Sin Nombre/genética , Vírus Sin Nombre/patogenicidade , Vírus Sin Nombre/fisiologia , Tropismo , Células Vero , Virulência/genética , Replicação Viral
6.
J Virol ; 78(9): 4433-43, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15078924

RESUMO

Two decades after a worldwide vaccination campaign was used to successfully eradicate naturally occurring smallpox, the threat of bioterrorism has led to renewed vaccination programs. In addition, sporadic outbreaks of human monkeypox in Africa and a recent outbreak of human monkeypox in the U.S. have made it clear that naturally occurring zoonotic orthopoxvirus diseases remain a public health concern. Much of the threat posed by orthopoxviruses could be eliminated by vaccination; however, because the smallpox vaccine is a live orthopoxvirus vaccine (vaccinia virus) administered to the skin, the vaccine itself can pose a serious health risk. Here, we demonstrate that rhesus macaques vaccinated with a DNA vaccine consisting of four vaccinia virus genes (L1R, A27L, A33R, and B5R) were protected from severe disease after an otherwise lethal challenge with monkeypox virus. Animals vaccinated with a single gene (L1R) which encodes a target of neutralizing antibodies developed severe disease but survived. This is the first demonstration that a subunit vaccine approach to smallpox-monkeypox immunization is feasible.


Assuntos
Monkeypox virus/patogenicidade , Mpox/prevenção & controle , Vacina Antivariólica/administração & dosagem , Vacinas de DNA/administração & dosagem , Vaccinia virus/imunologia , Proteínas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Humanos , Macaca mulatta , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Vacina Antivariólica/imunologia , Vacinação , Vacinas de DNA/imunologia , Vaccinia virus/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Proteínas Virais/genética
7.
J Virol ; 77(18): 9894-905, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12941899

RESUMO

Hantavirus pulmonary syndrome (HPS) is a rapidly progressing human disease with one of the highest case fatality rates (30 to 50%) of any acute viral disease known. There are no vaccines, effective antiviral drugs, or immunologics to prevent or treat HPS. In an attempt to develop HPS medical countermeasures, we constructed an expression plasmid, pWRG/AND-M, that contains the full-length M genome segment of Andes virus (ANDV), a South American hantavirus. Transfection experiments in cell culture indicated that both the G1 and G2 glycoproteins are expressed from pWRG/AND-M. Rhesus macaques vaccinated by gene gun with pWRG/AND-M developed remarkably high levels of neutralizing antibodies that not only neutralized ANDV but also cross-neutralized other HPS-associated hantaviruses, including Sin Nombre virus. To determine if the antibodies elicited in the monkeys could confer protection, we performed a series of passive-transfer experiments using a recently described lethal HPS animal model (i.e., adult Syrian hamsters develop HPS and die within 10 to 15 days after challenge with ANDV). When injected into hamsters 1 day before challenge, sera from the vaccinated monkeys either provided sterile protection or delayed the onset of HPS and death. When injected on day 4 or 5 after challenge, the monkey sera protected 100% of the hamsters from lethal disease. These data provide a proof of concept for a gene-based HPS vaccine and also demonstrate the potential value of a postexposure immunoprophylactic to treat individuals after exposure, or potential exposure, to these highly lethal hantaviruses.


Assuntos
Síndrome Pulmonar por Hantavirus/prevenção & controle , Orthohantavírus/imunologia , Vacinas de DNA/imunologia , Vacinas Virais/imunologia , Animais , Células COS , Cricetinae , Feminino , Genoma Viral , Orthohantavírus/genética , Humanos , Imunização Passiva , Macaca mulatta , Mesocricetus , Vacinação
9.
Virology ; 289(1): 6-14, 2001 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-11601912

RESUMO

Hantaviruses are associated with two human diseases, hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS). Development of vaccines and therapies to prevent and treat HFRS and HPS have been hampered by the absence of a practical animal model. Here we report that Andes virus (ANDV), a South American hantavirus, is highly lethal in adult Syrian hamsters. The characteristics of the disease in hamsters, including the incubation period, symptoms of rapidly progressing respiratory distress, and pathologic findings of pulmonary edema and pleural effusion, closely resemble HPS in humans. This is the first report of a lethal disease model for hantaviruses that causes HPS.


Assuntos
Modelos Animais de Doenças , Síndrome Pulmonar por Hantavirus , Mesocricetus , Orthohantavírus/patogenicidade , Animais , Cricetinae , Feminino , Orthohantavírus/isolamento & purificação , Síndrome Pulmonar por Hantavirus/mortalidade , Síndrome Pulmonar por Hantavirus/patologia , Síndrome Pulmonar por Hantavirus/fisiopatologia , Síndrome Pulmonar por Hantavirus/virologia , Humanos , Pulmão/patologia , Pulmão/virologia
10.
Protein Expr Purif ; 23(1): 134-41, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11570855

RESUMO

Sin Nombre virus is a member of the Hantavirus genus, family Bunyaviridae, and is an etiologic agent of hantavirus pulmonary syndrome. The hantavirus nucleocapsid (N) protein plays an important role in the encapsidation and assembly of the viral negative-sense genomic RNA. The Sin Nombre N protein was expressed as a C-terminal hexahistidine fusion in Escherichia coli and initially purified by nickel-affinity chromatography. We developed methods to extract the soluble fraction and to solubilize the remainder of the N protein using denaturants. Maximal expression of protein from native purification was observed after a 1.5-h induction with IPTG (2.4 mg/L). The zwitterionic detergent Chaps did not enhance the yield of native purifications, but increased the yield of protein obtained from insoluble purifications. Both soluble and insoluble materials, purified by nickel-affinity chromatography, were also subjected to Hi Trap SP Sepharose fast-flow (FF) chromatography. Both soluble and insoluble proteins had a similar A(280) profile on the Sepharose FF column, and both suggested the presence of a nucleic acid contaminant. The apparent dissociation constant of the N protein, purified by nickel-affinity and SP Sepharose FF chromatography, and the 5' end of the viral S-segment genome were measured using a filter binding assay. The N protein-vRNA complex had an apparent dissociation constant of 140 nM.


Assuntos
Escherichia coli/virologia , Proteínas do Nucleocapsídeo/isolamento & purificação , Vírus Sin Nombre/química , Marcadores de Afinidade , Ácidos Cólicos/farmacologia , Cromatografia , Cromatografia de Afinidade , Detergentes/farmacologia , Histidina , Proteínas do Nucleocapsídeo/biossíntese , Proteínas do Nucleocapsídeo/metabolismo , Ligação Proteica , Desnaturação Proteica , RNA/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo
11.
J Virol ; 75(18): 8469-77, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11507192

RESUMO

Four hantaviruses-Hantaan virus (HTNV), Seoul virus (SEOV), Dobrava virus (DOBV) and Puumala virus-are known to cause hemorrhagic fever with renal syndrome (HFRS) in Europe and Asia. HTNV causes the most severe form of HFRS (5 to 15% case-fatality rate) and afflicts tens of thousands of people annually. Previously, we demonstrated that DNA vaccination with a plasmid expressing the SEOV M gene elicited neutralizing antibodies and protected hamsters against infection with SEOV and HTNV. Here, we report the construction and evaluation of a DNA vaccine that expresses the HTNV M gene products, G1 and G2. DNA vaccination of hamsters with the HTNV M gene conferred sterile protection against infection with HTNV, SEOV, and DOBV. DNA vaccination of rhesus monkeys with either the SEOV or HTNV M gene elicited high levels of neutralizing antibodies. These are the first immunogenicity data for hantavirus DNA vaccines in nonhuman primates. Because a neutralizing antibody response is considered a surrogate marker for protective immunity in humans, our protection data in hamsters combined with the immunogenicity data in monkeys suggest that hantavirus M gene-based DNA vaccines could protect humans against the most severe forms of HFRS.


Assuntos
Anticorpos Antivirais/imunologia , DNA Viral/imunologia , Vírus Hantaan/imunologia , Febre Hemorrágica com Síndrome Renal/prevenção & controle , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Antivirais/biossíntese , Células COS , Chlorocebus aethiops , Cricetinae , Reações Cruzadas , Modelos Animais de Doenças , Expressão Gênica , Genes Virais , Vírus Hantaan/genética , Vírus Hantaan/isolamento & purificação , Febre Hemorrágica com Síndrome Renal/virologia , Macaca mulatta , Testes de Neutralização , Primatas , Vacinação , Vacinas de DNA , Proteínas do Envelope Viral/genética
13.
Trends Microbiol ; 8(2): 61-7, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10664598

RESUMO

Hantaviruses include serious human pathogens that are maintained in nature in persistently infected rodents and that can also persistently infect cultured mammalian cells, causing little or no cytopathology. The mechanisms of hantavirus persistence are only beginning to be explored. Recent data point to subtle changes in the viral genome that might result in the differential regulation of replication and lead to persistence.


Assuntos
Infecções por Hantavirus/veterinária , Orthohantavírus/fisiologia , Doenças dos Roedores/virologia , Animais , Reservatórios de Doenças , Genoma Viral , Vírus Hantaan/fisiologia , Orthohantavírus/genética , Infecções por Hantavirus/virologia , Humanos , Replicação Viral
14.
Virology ; 266(2): 329-39, 2000 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-10639319

RESUMO

Previously we found that passive transfer of monoclonal antibodies (MAbs) specific to either the vaccinia virus (VACV) L1R or A33R gene product protected mice from challenge with VACV. The L1R-specific MAbs, which bind the intracellular mature virion (IMV), neutralized virus in cell culture, whereas the A33R-specific MAbs, which bind extracellular enveloped virions (EEV), did not. To investigate whether a protective response could be generated by vaccination with these genes, we constructed and evaluated DNA vaccines expressing the VACV L1R and/or A33R genes under control of a cytomegalovirus promoter. Mice were vaccinated with DNA-coated gold beads by using a gene gun and then challenged with VACV (strain WR) intraperitoneally. Mice vaccinated with L1R alone developed neutralizing antibodies and were partially protected. Mice vaccinated with a combination of both genes loaded on the same gold beads developed a robust anti-A33R response; however, no neutralizing antibody response was detected, and the mice were not protected. In contrast, when mice were vaccinated with L1R and A33R loaded on different gold beads, neutralizing (presumably anti-L1R) and anti-A33R antibody responses were detected, and protection was markedly improved. Our results indicated that vaccination with both L1R and A33R proteins, intended to evoke mechanistically distinct and complementary forms of protection, was more effective than vaccination with either protein by itself.


Assuntos
Infecções por Poxviridae/prevenção & controle , Vacinas de DNA/farmacologia , Vaccinia virus/genética , Vacinas Virais/farmacologia , Animais , Anticorpos Antivirais/biossíntese , Células COS , Clonagem Molecular , Genes Virais , Camundongos , Testes de Neutralização , Poxviridae/patogenicidade , Vaccinia virus/imunologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
15.
J Med Virol ; 60(1): 77-85, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10568767

RESUMO

We evaluated a vaccinia-vectored vaccine for hemorrhagic fever with renal syndrome in clinical trials. A Phase I dose-escalation study in 16 volunteers divided into four groups demonstrated that subcutaneous inoculation of approximately 10(7) plaque-forming units of the recombinant virus was safe and immunogenic. Vaccination of a fifth group of 12 volunteers indicated that neutralizing antibody titers to both vaccinia virus and Hantaan virus were enhanced after a second inoculation. Comparing two routes of vaccination showed that scarification effectively induced neutralizing antibodies in vaccinia virus-naive volunteers but that subcutaneous inoculation was superior to scarification in vaccinia virus-immune individuals. A Phase II, double-blinded, placebo-controlled clinical trial was conducted among 142 volunteers. Two subcutaneous vaccinations were administered at 4-week intervals. Neutralizing antibodies to Hantaan virus or to vaccinia virus were detected in 72% or 98% of vaccinia virus-naive volunteers, respectively. In contrast, only 26% of the vaccinia virus-immune volunteers developed neutralizing antibody responses to Hantaan virus. J. Med. Virol. 60:77-85, 2000. Published 2000 Wiley-Liss, Inc.


Assuntos
Anticorpos Antivirais/sangue , Vírus Hantaan/imunologia , Vacinas Sintéticas/imunologia , Vaccinia virus/genética , Vacinas Virais/imunologia , Relação Dose-Resposta Imunológica , Método Duplo-Cego , Ensaio de Imunoadsorção Enzimática , Vetores Genéticos , Vírus Hantaan/genética , Humanos , Imunização Secundária , Ativação Linfocitária , Testes de Neutralização , Vacinação , Vacinas Sintéticas/administração & dosagem , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
16.
J Biol Chem ; 274(47): 33732-9, 1999 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-10559265

RESUMO

The nucleocapsid (N) protein functions in hantavirus replication through its interactions with the viral genomic and antigenomic RNAs. To address the biological functions of the N protein, it was critical to first define this binding interaction. The dissociation constant, K(d), for the interaction of the Hantaan virus (HTNV) N protein and its genomic S segment (vRNA) was measured under several solution conditions. Overall, increasing the NaCl and Mg(2+) in these binding reactions had little impact on the K(d). However, the HTNV N protein showed an enhanced specificity for HTNV vRNA as compared with the S segment open reading frame RNA or a nonviral RNA with increasing ionic strength and the presence of Mg(2+). In contrast, the assembly of Sin Nombre virus N protein-HTNV vRNA complexes was inhibited by the presence of Mg(2+) or an increase in the ionic strength. The K(d) values for HTNV and Sin Nombre virus N proteins were nearly identical for the S segment open reading frame RNA, showing weak affinity over several binding reaction conditions. Our data suggest a model in which specific recognition of the HTNV vRNA by the HTNV N protein resides in the noncoding regions of the HTNV vRNA.


Assuntos
Proteínas do Capsídeo , Capsídeo/metabolismo , Vírus Hantaan/metabolismo , RNA Viral/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas do Core Viral/metabolismo , Capsídeo/isolamento & purificação , Magnésio/metabolismo , Concentração Osmolar , Ligação Proteica , Proteínas do Core Viral/isolamento & purificação
17.
Virology ; 263(1): 209-19, 1999 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-10544095

RESUMO

Seoul virus (SEOV) is a member of the Hantavirus genus (family Bunyaviridae) and an etiological agent of hemorrhagic fever with renal syndrome. The medium (M) and small (S) gene segments of SEOV encode the viral envelope glycoproteins and nucleocapsid protein, respectively. We compared the immunogenicity and protective efficacy of naked DNA (pWRG7077), DNA-based Sindbis replicon (pSIN2.5), and packaged Sindbis replicon vectors (pSINrep5), containing either the M or S gene segment of SEOV in Syrian hamsters. All of the vectors elicited an anti-SEOV immune response to the expressed SEOV gene products. Vaccinated hamsters were challenged with SEOV and monitored for evidence of infection. Protection from infection was strongly associated with M-gene vaccination. A small number of S-gene-vaccinated animals also were protected. Hamsters vaccinated with the pWRG7077 vector expressing the M gene demonstrated the most consistent protection from SEOV infection and also were protected from heterologous hantavirus (Hantaan virus) infection.


Assuntos
Infecções por Hantavirus/prevenção & controle , Orthohantavírus/imunologia , Vacinas de DNA/imunologia , Proteínas Estruturais Virais/imunologia , Vacinas Virais/imunologia , Animais , Cricetinae , DNA Viral/imunologia , Ensaio de Imunoadsorção Enzimática , Vetores Genéticos , Orthohantavírus/genética , Infecções por Hantavirus/imunologia , Mesocricetus , Testes de Neutralização , Proteínas do Nucleocapsídeo/genética , Testes de Precipitina , Replicon/imunologia , Sindbis virus/imunologia , Vacinas de DNA/administração & dosagem , Proteínas do Envelope Viral/genética , Proteínas Estruturais Virais/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
18.
J Virol ; 73(7): 5301-8, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10364276

RESUMO

Hantaan virus, the prototypic member of the Hantavirus genus, causes hemorrhagic fever with renal syndrome in humans. We examined the human memory T-lymphocyte responses of three donors who had previous laboratory-acquired infections with Hantaan virus. We demonstrated virus-specific responses in bulk cultures of peripheral blood mononuclear cells (PBMC) from all donors. Bulk T-cell responses were directed against either Hantaan virus nucleocapsid (N) or G1 protein, and these responses varied between donors. We established both CD4(+) and CD8(+) N-specific cell lines from two donors and CD4(+) G1-specific cell lines from a third donor. All CD8(+) cytotoxic T-lymphocyte (CTL) lines recognized one of two epitopes on the nucleocapsid protein: one epitope spanning amino acids 12 to 20 and the other spanning amino acids 421 to 429. The CTL lines specific for amino acids 12 to 20 were restricted by HLA B51, and those specific for amino acids 421 to 429 were restricted by HLA A1. The N-specific CTL lines isolated from these two donors included both Hantaan virus-specific CTLs and hantavirus cross-reactive CTLs. Responses to both epitopes are detectable in short-term bulk cultures of PBMC from one donor, and precursor frequency analysis confirms that CTLs specific for these epitopes are present at relatively high precursor frequencies in the peripheral T-cell pool. These data suggest that infection with Hantaan virus results in the generation of CTL to limited epitopes on the nucleocapsid protein and that infection also results in the generation of cross-reactive T-cell responses to distantly related hantaviruses which cause the distinct hantavirus pulmonary syndrome. This is the first demonstration of human T-lymphocyte responses to Hantaan virus.


Assuntos
Proteínas do Capsídeo , Capsídeo/imunologia , Epitopos de Linfócito T/imunologia , Vírus Hantaan/imunologia , Febre Hemorrágica com Síndrome Renal/imunologia , Memória Imunológica/imunologia , Linfócitos T Citotóxicos/imunologia , Proteínas do Core Viral/imunologia , Sequência de Aminoácidos , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Células Cultivadas , Reações Cruzadas , Antígenos HLA-A/imunologia , Antígenos HLA-B/imunologia , Humanos , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/imunologia , Dados de Sequência Molecular , Peptídeos/imunologia
19.
Virology ; 255(2): 269-78, 1999 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-10069952

RESUMO

Seoul virus (SEOV) is one of four known hantaviruses causing hemorrhagic fever with renal syndrome (HFRS). Candidate naked DNA vaccines for HFRS were constructed by subcloning cDNA representing the medium (M; encoding the G1 and G2 glycoproteins) or small (S; encoding the nucleocapsid protein) genome segment of SEOV into the DNA expression vector pWRG7077. We vaccinated BALB/c mice with three doses of the M or S DNA vaccine at 4-week intervals by either gene gun inoculation of the epidermis or needle inoculation into the gastrocnemius muscle. Both routes of vaccination resulted in antibody responses as measured by ELISA; however, gene gun inoculation elicited a higher frequency of seroconversion and higher levels of antibodies in individual mice. We vaccinated Syrian hamsters with the M or S construct using the gene gun and found hantavirus-specific antibodies in five of five and four of five hamsters, respectively. Animals vaccinated with the M construct developed a neutralizing antibody response that was greatly enhanced in the presence of guinea pig complement. Immunized hamsters were challenged with SEOV and, after 28 days, were monitored for evidence of infection. Hamsters vaccinated with M were protected from infection, but hamsters vaccinated with S were not protected.


Assuntos
Anticorpos Antivirais/imunologia , Febre Hemorrágica com Síndrome Renal/prevenção & controle , Proteínas do Nucleocapsídeo/imunologia , Vacinas de DNA/imunologia , Vacinas Sintéticas/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Antivirais/biossíntese , Células COS , Cricetinae , Expressão Gênica , Cobaias , Orthohantavírus/genética , Orthohantavírus/imunologia , Febre Hemorrágica com Síndrome Renal/imunologia , Mesocricetus , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Proteínas do Nucleocapsídeo/genética , Vacinação , Proteínas do Envelope Viral/genética
20.
Clin Diagn Virol ; 8(2): 113-21, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9316733

RESUMO

BACKGROUND: Hantaviruses cause two serious human diseases: hantavirus pulmonary syndrome and hemorrhagic fever with renal syndrome. At least nine hantaviruses are known to be pathogenic for humans and numerous others, with unknown disease potential, have been detected in rodents. Assays to quickly identify specific hantaviruses would be useful both for clinical diagnosis and in risk assessment studies. OBJECTIVES: The goal of our study was to develop and test a specific and sensitive PCR-based assay for identification and differentiation of hantaviruses. STUDY DESIGN: We developed an assay that combined RNA-PCR amplification and colorimetric enzymatic detection to identify representative European, Asian, and north American hantaviruses. RNAs from 18 hantavirus strains of nine species were amplified in the presence of digoxigenin-dUTP by using a single pair of oligonucleotide primers and polymerase chain reaction (PCR) performed by using rTth DNA polymerase. Digoxigenin-labeled PCR products were hybridized in solution to virus type-specific biotinilated probes, captured onto streptavidin-coated microtiter plates and detected by horseradish peroxidase-labeled anti-digoxigenin antibodies and a chromogenic substrate. RESULTS AND CONCLUSIONS: The assay correctly identified each homologous virus type tested. The detection limit of the assay was approximately 15 PFU or at least 50 copies of the viral genome. The assay is simple and strain-specific and is adaptable for automation, making it more practical than other available techniques for accurate and reliable diagnosis and typing of hantaviruses.


Assuntos
Infecções por Hantavirus/diagnóstico , Orthohantavírus/isolamento & purificação , Reação em Cadeia da Polimerase/métodos , RNA Viral/análise , Infecções por Hantavirus/virologia , Humanos , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA