Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Clin Microbiol Infect ; 20 Suppl 5: 110-7, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24447183

RESUMEN

Haemorrhagic fever with renal syndrome (HFRS) is endemic in Asia, Europe and Scandinavia, and is caused by infection with the hantaviruses Hantaan (HTNV), Seoul (SEOV), Puumala (PUUV), or Dobrava (DOBV) viruses. We developed candidate DNA vaccines for HFRS expressing the Gn and Gc genes of HTNV or PUUV and evaluated them in an open-label, single-centre Phase 1 study. Three groups of nine participants each were vaccinated on days 0, 28 and 56 with the DNA vaccines for HTNV, PUUV, or a mixture of both vaccines using the Ichor Medical Systems TriGrid Intramuscular Delivery System. All vaccinations consisted of a total dose of 2.0 mg DNA in an injected volume of 1 mL saline. For the combined vaccine, the mixture contained equal amounts (1.0 mg) of each DNA vaccine. There were no study-related serious adverse events. Neutralizing antibody responses were measured by a plaque reduction neutralization test. Neutralizing antibody responses were detected in five of nine and seven of nine individuals who completed all three vaccinations with the HTNV or PUUV DNA vaccines, respectively. In the combined vaccine group, seven of the nine volunteers receiving all three vaccinations developed neutralizing antibodies to PUUV. The three strongest responders to the PUUV vaccine also had strong neutralizing antibody responses to HTNV. These results demonstrate that the HTNV and PUUV DNA vaccines delivered by electroporation separately or as a mixture are safe. In addition, both vaccines were immunogenic, although when mixed together, more participants responded to the PUUV than to the HTNV DNA vaccine.


Asunto(s)
Virus Hantaan/genética , Fiebre Hemorrágica con Síndrome Renal/prevención & control , Virus Puumala/genética , Vacunas de ADN/administración & dosificación , Vacunas Virales/administración & dosificación , Adolescente , Adulto , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Electroporación , Femenino , Humanos , Inyecciones Intramusculares , Masculino , Persona de Mediana Edad , Pruebas de Neutralización , Vacunación , Vacunas de ADN/efectos adversos , Vacunas de ADN/inmunología , Vacunas Virales/efectos adversos , Adulto Joven
2.
Clin Vaccine Immunol ; 20(2): 218-26, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23239797

RESUMEN

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.


Asunto(s)
Infecciones por Hantavirus/inmunología , Fiebre Hemorrágica con Síndrome Renal/inmunología , Virus Puumala/inmunología , Vacunas de ADN/inmunología , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Células COS , Línea Celular , Chlorocebus aethiops , Cricetinae , Reacciones Cruzadas , ADN Viral/inmunología , Orthohantavirus/inmunología , Fiebre Hemorrágica con Síndrome Renal/prevención & control , Fiebre Hemorrágica con Síndrome Renal/virología , Macaca mulatta/inmunología , Pruebas de Neutralización , Vacunación , Vacunas de ADN/administración & dosificación , Células Vero , Ensayo de Placa Viral , Vacunas Virales/administración & dosificación
3.
Vaccine ; 29(39): 6728-35, 2011 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-21219978

RESUMEN

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.


Asunto(s)
Citometría de Flujo/métodos , Orthohantavirus/inmunología , Vacunas de ADN/inmunología , Animales , Antígenos Virales/inmunología , Biolística , Células COS , Chlorocebus aethiops , Sistemas de Liberación de Medicamentos/métodos , Estabilidad de Medicamentos , Electroforesis en Gel de Agar , Electroporación , Virus de la Encefalitis Equina Venezolana/genética , Virus de la Encefalitis Equina Venezolana/inmunología , Citometría de Flujo/instrumentación , Orthohantavirus/genética , Humanos , Plásmidos/genética , Plásmidos/metabolismo , Estándares de Referencia , Reproducibilidad de los Resultados , Transfección , Vacunas de ADN/administración & dosificación , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología
4.
Virology ; 326(1): 130-9, 2004 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-15262501

RESUMEN

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.


Asunto(s)
Genoma Viral , Infecciones por Hantavirus/virología , Orthohantavirus/fisiología , Virus Reordenados/fisiología , Animales , Chlorocebus aethiops , Cricetinae , Modelos Animales de Enfermedad , Femenino , Orthohantavirus/genética , Orthohantavirus/patogenicidad , Mesocricetus , Virus Reordenados/patogenicidad , Virus Sin Nombre/genética , Virus Sin Nombre/patogenicidad , Virus Sin Nombre/fisiología , Tropismo , Células Vero , Virulencia/genética , Replicación Viral
5.
J Virol ; 78(9): 4433-43, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15078924

RESUMEN

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.


Asunto(s)
Monkeypox virus/patogenicidad , Mpox/prevención & control , Vacuna contra Viruela/administración & dosificación , Vacunas de ADN/administración & dosificación , Virus Vaccinia/inmunología , Proteínas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Humanos , Macaca mulatta , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Vacuna contra Viruela/inmunología , Vacunación , Vacunas de ADN/inmunología , Virus Vaccinia/genética , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Proteínas Virales/genética
6.
J Virol ; 77(18): 9894-905, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12941899

RESUMEN

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.


Asunto(s)
Síndrome Pulmonar por Hantavirus/prevención & control , Orthohantavirus/inmunología , Vacunas de ADN/inmunología , Vacunas Virales/inmunología , Animales , Células COS , Cricetinae , Femenino , Genoma Viral , Orthohantavirus/genética , Humanos , Inmunización Pasiva , Macaca mulatta , Mesocricetus , Vacunación
7.
Virology ; 306(1): 181-95, 2003 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-12620810

RESUMEN

Two major infectious forms of vaccinia virus (VACV) have been described: the intracellular mature virion (IMV), and the extracellular enveloped virion (EEV). Due to their stability in the environment, IMVs play a predominant role in host-to-host transmission, whereas EEVs play an important role in dissemination within the host. In a previous report, we demonstrated that mice vaccinated with VACV L1R (IMV immunogen) and A33R (EEV immunogen) were protected from a lethal poxvirus challenge. Vaccination with a combination of both genes conferred greater protection than either gene alone, suggesting that an immune response against both IMV and EEV is advantageous. Here, we report that in mice individually administered DNA vaccines with two different VACV immunogens, A27L (IMV immunogen) or B5R (EEV immunogen), failed to significantly protect; however, vaccination with a combination of both genes conferred a high level of protection. Mice were completely protected when vaccinated with a combination of four VACV genes (A27L + A33R + L1R + B5R). Rhesus macaques vaccinated with this four-gene-combination developed appropriate antibody responses to each protein. Antibody responses elicited by this vaccine cross-reacted with monkeypox virus orthologous proteins. These data indicate that a gene-based vaccine comprised of the VACV A27L + A33R + L1R + B5R genes may be a useful candidate to protect against other orthopoxviruses, including those that cause monkeypox and smallpox.


Asunto(s)
Anticuerpos Antivirales/sangre , Vacunas de ADN/inmunología , Virus Vaccinia/inmunología , Vaccinia/prevención & control , Proteínas Virales/inmunología , Vacunas Virales/inmunología , Secuencia de Aminoácidos , Animales , Humanos , Macaca mulatta , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Datos de Secuencia Molecular , Plásmidos/genética , Vacunas de ADN/administración & dosificación , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Proteínas Virales/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
8.
Virology ; 289(1): 6-14, 2001 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-11601912

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Síndrome Pulmonar por Hantavirus , Mesocricetus , Orthohantavirus/patogenicidad , Animales , Cricetinae , Femenino , Orthohantavirus/aislamiento & purificación , Síndrome Pulmonar por Hantavirus/mortalidad , Síndrome Pulmonar por Hantavirus/patología , Síndrome Pulmonar por Hantavirus/fisiopatología , Síndrome Pulmonar por Hantavirus/virología , Humanos , Pulmón/patología , Pulmón/virología
9.
J Virol ; 75(18): 8469-77, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11507192

RESUMEN

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.


Asunto(s)
Anticuerpos Antivirales/inmunología , ADN Viral/inmunología , Virus Hantaan/inmunología , Fiebre Hemorrágica con Síndrome Renal/prevención & control , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Antivirales/biosíntesis , Células COS , Chlorocebus aethiops , Cricetinae , Reacciones Cruzadas , Modelos Animales de Enfermedad , Expresión Génica , Genes Virales , Virus Hantaan/genética , Virus Hantaan/aislamiento & purificación , Fiebre Hemorrágica con Síndrome Renal/virología , Macaca mulatta , Pruebas de Neutralización , Primates , Vacunación , Vacunas de ADN , Proteínas del Envoltorio Viral/genética
11.
Virology ; 266(2): 329-39, 2000 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-10639319

RESUMEN

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.


Asunto(s)
Infecciones por Poxviridae/prevención & control , Vacunas de ADN/farmacología , Virus Vaccinia/genética , Vacunas Virales/farmacología , Animales , Anticuerpos Antivirales/biosíntesis , Células COS , Clonación Molecular , Genes Virales , Ratones , Pruebas de Neutralización , Poxviridae/patogenicidad , Virus Vaccinia/inmunología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
12.
Virology ; 263(1): 209-19, 1999 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-10544095

RESUMEN

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.


Asunto(s)
Infecciones por Hantavirus/prevención & control , Orthohantavirus/inmunología , Vacunas de ADN/inmunología , Proteínas Estructurales Virales/inmunología , Vacunas Virales/inmunología , Animales , Cricetinae , ADN Viral/inmunología , Ensayo de Inmunoadsorción Enzimática , Vectores Genéticos , Orthohantavirus/genética , Infecciones por Hantavirus/inmunología , Mesocricetus , Pruebas de Neutralización , Proteínas de la Nucleocápside/genética , Pruebas de Precipitina , Replicón/inmunología , Virus Sindbis/inmunología , Vacunas de ADN/administración & dosificación , Proteínas del Envoltorio Viral/genética , Proteínas Estructurales Virales/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
13.
Virology ; 255(2): 269-78, 1999 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-10069952

RESUMEN

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.


Asunto(s)
Anticuerpos Antivirales/inmunología , Fiebre Hemorrágica con Síndrome Renal/prevención & control , Proteínas de la Nucleocápside/inmunología , Vacunas de ADN/inmunología , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Antivirales/biosíntesis , Células COS , Cricetinae , Expresión Génica , Cobayas , Orthohantavirus/genética , Orthohantavirus/inmunología , Fiebre Hemorrágica con Síndrome Renal/inmunología , Mesocricetus , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización , Proteínas de la Nucleocápside/genética , Vacunación , Proteínas del Envoltorio Viral/genética
14.
J Virol ; 70(1): 672-7, 1996 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8523592

RESUMEN

Reovirus intermediate subviral particles (ISVPs) but not intact virions or cores have been shown to possess the capacity to permeabilize mouse L cells as determined by a 51Cr release assay. We used monoclonal antibodies (MAbs) directed against proteins exposed on the ISVP surface (sigma 1, mu 1, and lambda 2) to probe the role(s) of these proteins in membrane interaction and penetration. One sigma 1-specific MAb (MAb-G5) and two mu 1-specific MAbs (MAb-10H2 and MAb-8H6) inhibited reovirus-induced 51Cr release when added pre- or post-ISVP attachment to L cells. MAb-G5 inhibits 51Cr release by interfering with ISVP attachment (via sigma 1) to L-cell receptor sites. The mu 1-specific MAbs (MAb-10H2 and MAb-8H6) inhibit 51Cr release by interfering with an undefined post-L-cell-attachment event that involves bivalent binding of the mu 1-specific MAbs to an epitope located in a central region of the mu 1 protein.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Proteínas de la Cápside , Cápside/inmunología , Cromo/metabolismo , Proteínas Virales/inmunología , Animales , Permeabilidad de la Membrana Celular , Mapeo Epitopo , Fibroblastos/citología , Células L , Ratones
15.
J Virol ; 70(1): 459-67, 1996 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8523558

RESUMEN

The reovirus M2 gene is associated with the capacity of type 3 strain Abney (T3A) intermediate subviral particles (ISVPs) to permeabilize cell membranes as measured by chromium (51Cr) release (P. Lucia-Jandris, J. W. Hooper, and B. N. Fields, J. Virol. 67:5339-5345, 1993). In addition, reovirus mutants with lesions in the M2 gene can be selected by heating virus at 37 degrees C for 20 min in 33% ethanol (D. R. Wessner and B. N. Fields, J. Virol. 67:2442-2447, 1993). In this report we investigated the mechanism by which the reovirus M2 gene product (the mu 1 protein) influences the capacity of reovirus ISVPs to permeabilize membranes, using ethanol-selected T3A mutants. Each of three T3A ethanol-resistant mutants isolated (JH2, JH3, and JH4) exhibited a decreased capacity to cause 51Cr release relative to that of wild-type T3A. Sequence analysis of the M2 genes of wild-type T3A and the T3A mutants indicated that each mutant possesses a single amino acid substitution in a central region of the 708-amino-acid mu 1 protein: JH2 (residue 466, Tyr to Cys), JH3 (residue 459, Lys to Glu), and JH4 (residue 497 Pro to Ser). Assays performed with reovirus natural isolates, reassortants, and a set of previously characterized type 3 strain Dearing (T3D) ethanol-resistant mutants revealed a strong correlation between ethanol sensitivity and the capacity to cause 51Cr release. We found that ISVPs generated from the T3A and T3D mutants were stable when heated to 50 degrees C, whereas wild-type T3A ISVPs are inactivated under these conditions. Together, these data suggest that amino acid substitutions in a central region of the mu 1 protein affect the capacity of the ISVP to permeabilize L-cell membranes by altering the stability of the virus particle.


Asunto(s)
Proteínas de la Cápside , Cápside/metabolismo , Cromo/metabolismo , Orthoreovirus Mamífero 3/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Cápside/genética , Permeabilidad de la Membrana Celular , Radioisótopos de Cromo/metabolismo , ADN Viral , Resistencia a Medicamentos , Etanol/farmacología , Genes Virales , Calor , Células L , Orthoreovirus Mamífero 3/efectos de los fármacos , Orthoreovirus Mamífero 3/genética , Ratones , Datos de Secuencia Molecular , Fenotipo , Virus Reordenados , Supresión Genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
16.
J Pharm Biomed Anal ; 13(2): 89-97, 1995 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-7766728

RESUMEN

Although some degree of consensus has been reached concerning the requirements for acceptable method validation, the procedures used to establish them vary significantly between laboratories. Also, issues arising from application of these requirements during validation and subsequent sample analysis need to be addressed. The purpose of this paper is to discuss application issues concerning prerequisites to method validation, and all validation criteria for evaluation of method reliability and overall performance. Other poorly addressed issues such as re-validation, cross-validation, partial sample volume, multicomponent analysis and reporting will also be discussed. Although many issues discussed are of a general nature, the scope of this presentation is primarily to address issues arising from the validation and routine application of chromatographic methods.


Asunto(s)
Reproducibilidad de los Resultados , Animales , Cromatografía/métodos , Cromatografía/normas , Interpretación Estadística de Datos , Humanos , Farmacocinética
17.
J Virol ; 67(9): 5339-45, 1993 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8350400

RESUMEN

In this study, we investigated the interaction of reovirus particles with cell membranes by using a 51Cr release assay. We confirmed prior observations (J. Borsa, B. D. Morash, M. D. Sargent, T. P. Copps, P. A. Lievaart, and J. G. Szekely, J. Gen. Virol. 45:161-170, 1979) that intermediate subviral particles (ISVPs) of reovirus type 3 strain Abney (T3A) induced the release of 51Cr from preloaded L cells and showed that the intact virion and core forms did not. Reovirus type 1 strain Lang (T1L) ISVPs were found to be less efficient at 51Cr release than T3A ISVPs. Reassortants between these strains indicated that the 51Cr release phenotype segregates with the M2 gene segment. Biochemical studies indicated that the ISVPs' acquisition of the capacity to induce 51Cr release followed the cleavage of the viral M2 gene product mu 1/mu 1C to fragments delta and phi during virion conversion to ISVP but did not directly correlate with this cleavage. These studies suggest that the reovirus M2 gene product (in its cleaved form) plays a role in interacting with cell membranes.


Asunto(s)
Transformación Celular Viral , Cromo/metabolismo , Genes Virales , Reoviridae/genética , Animales , Membrana Celular/metabolismo , Quimotripsina/metabolismo , Quimotripsina/farmacología , Cinética , Células L , Orthoreovirus Mamífero 3/genética , Orthoreovirus Mamífero 3/fisiología , Ratones , Reoviridae/efectos de los fármacos , Reoviridae/fisiología , Factores de Tiempo , Virión/genética , Virión/fisiología
18.
Clin Pharmacol Ther ; 39(3): 300-5, 1986 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-3948468

RESUMEN

Oral ranitidine was given to 68 healthy subjects between 18 and 75 years old at a dosage of 150 mg twice a day for seven doses. Fifteen subjects were 18 to 35 years old (group I), 19 subjects were 36 to 50 years old (group II), 19 subjects were 51 to 65 years old (group III), and 15 subjects were 66 to 75 years old (group IV). Venous blood samples were drawn and the AUC from 0 to 12 hours, the maximum plasma concentration, the time of the maximum plasma concentration, the minimum plasma concentration, and the elimination t1/2 were determined. When groups III and IV were compared with groups I or II, significant (P less than 0.05) increases were seen in the AUC(0-12) (42% and 50%), the maximum plasma concentration (36% and 41%), the minimum plasma concentration (91% and 85%), and the elimination t1/2 (29% and 33%). Positive linear correlations were found when the AUC(0-12) (r = 0.68; P less than 0.01), the maximum plasma concentration (r = 0.34; P less than 0.01), the minimum plasma concentration (r = 0.55; P less than 0.01), and the elimination t1/2 (r = 0.46; P less than 0.01) were regressed with age. Our results suggest that it may be appropriate to consider dosage adjustments for patients over 50 years of age who take ranitidine.


Asunto(s)
Envejecimiento , Ranitidina/metabolismo , Administración Oral , Adolescente , Adulto , Anciano , Análisis de Varianza , Cromatografía Líquida de Alta Presión , Semivida , Humanos , Cinética , Masculino , Persona de Mediana Edad , Ranitidina/sangre
19.
Clin Pharmacol Ther ; 38(5): 533-7, 1985 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-3876907

RESUMEN

After an 11-day baseline period, groups of eight healthy men received isoxicam, 200 mg once a day, or piroxicam, 20 mg once a day, for 28 days. Fecal blood loss (FBL) was quantitated by the 51Cr-labeled erythrocyte method. FBLs for both isoxicam and piroxicam were only slightly higher than baseline for the first 3 drug dosing days, confirming the results of a previously published 4-day study of piroxicam, but FBL subsequently steadily increased, with maxima occurring for most subjects receiving isoxicam in 2 or 3 weeks and for most subjects receiving piroxicam in 3 or 4 weeks. This implies that dosing for 7 days or less, as is frequent in FBL studies of nonsteroidal anti-inflammatory drugs, may be insufficient to detect the peak drug-induced FBL. In week 4, FBL for both drugs was elevated approximately 0.6 ml/day over baseline. FBLs in week 4 were comparable to those reported for naproxen and less than those reported for indomethacin and acetylsalicylic acid. Statistical analyses of weekly plasma drug minimum concentrations suggest steady state was reached at week 2 for isoxicam and week 3 for piroxicam. There were large between-subject variations in steady-state plasma drug concentrations for both drugs. One subject in each drug group indulged in excessive alcohol consumption during the medication period, with a concomitant significant increase in FBL, which suggests exacerbation of the drug effect.


Asunto(s)
Antiinflamatorios/toxicidad , Hemorragia Gastrointestinal/inducido químicamente , Tiazinas/toxicidad , Adolescente , Adulto , Humanos , Masculino , Piroxicam , Tiazinas/sangre , Factores de Tiempo
20.
Pharmacology ; 30(1): 40-4, 1985.
Artículo en Inglés | MEDLINE | ID: mdl-3975255

RESUMEN

This study involved a randomized parallel groups comparison of the effects of aspirin formulated as enteric-coated granules (25 subjects) or as buffered tablets (26 subjects) with that of a lactose placebo (5 subjects), on the gastric and duodenal mucosa, as determined by endoscopic examination 2 h after a fasting single 975-mg dose. A grading scale of 0 (no damage) to 4 (severe damage) was used. The granule formulation produced a statistically significant (p less than 0.05) lower severity (mean 0.40 +/- 0.58 vs. 3.00 +/- 0.94) and incidence (36% of subjects vs. 100%) of gastric lesions than the buffered aspirin formulation. None of the lesions produced by the granule formulation or the placebo was considered clinically significant by the blinded endoscopist, whereas 17 subjects on the buffered formulation (65%) had clinically meaningful stomach damage. The incidence of duodenal lesions was minimal and comparable for the two formulations.


Asunto(s)
Aspirina/efectos adversos , Enfermedades Gastrointestinales/inducido químicamente , Adolescente , Adulto , Aspirina/administración & dosificación , Tampones (Química) , Femenino , Mucosa Gástrica/patología , Enfermedades Gastrointestinales/patología , Humanos , Mucosa Intestinal/patología , Masculino , Comprimidos Recubiertos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA