Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 230
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
J Virol ; 91(15)2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28539438

RESUMO

Palivizumab, a humanized murine monoclonal antibody that recognizes antigenic site II on both the prefusion (pre-F) and postfusion (post-F) conformations of the respiratory syncytial virus (RSV) F glycoprotein, is the only prophylactic agent approved for use for the treatment of RSV infection. However, its relatively low neutralizing potency and high cost have limited its use to a restricted population of infants at high risk of severe disease. Previously, we isolated a high-potency neutralizing antibody, 5C4, that specifically recognizes antigenic site Ø at the apex of the pre-F protein trimer. We compared in vitro and in vivo the potency and protective efficacy of 5C4 and the murine precursor of palivizumab, antibody 1129. Both antibodies were synthesized on identical murine backbones as either an IgG1 or IgG2a subclass and evaluated for binding to multiple F protein conformations, in vitro inhibition of RSV infection and propagation, and protective efficacy in mice. Although 1129 and 5C4 had similar pre-F protein binding affinities, the 5C4 neutralizing activity was nearly 50-fold greater than that of 1129 in vitro In BALB/c mice, 5C4 reduced the peak titers of RSV 1,000-fold more than 1129 did in both the upper and lower respiratory tracts. These data indicate that antibodies specific for antigenic site Ø are more efficacious at preventing RSV infection than antibodies specific for antigenic site II. Our data also suggest that site Ø-specific antibodies may be useful for the prevention or treatment of RSV infection and support the use of the pre-F protein as a vaccine antigen.IMPORTANCE There is no vaccine yet available to prevent RSV infection. The use of the licensed antibody palivizumab, which recognizes site II on both the pre-F and post-F proteins, is restricted to prophylaxis in neonates at high risk of severe RSV disease. Recommendations for using passive immunization in the general population or for therapy in immunocompromised persons with persistent infection is limited because of cost, determined from the high doses needed to compensate for its relatively low neutralizing potency. Prior efforts to improve the in vitro potency of site II-specific antibodies did not translate to significant in vivo dose sparing. We isolated a pre-F protein-specific, high-potency neutralizing antibody (5C4) that recognizes antigenic site Ø and compared its efficacy to that of the murine precursor of palivizumab (antibody 1129) matched for isotype and pre-F protein binding affinities. Our findings demonstrate that epitope specificity is an important determinant of antibody neutralizing potency, and defining the mechanisms of neutralization has the potential to identify improved products for the prevention and treatment of RSV infection.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Anticorpos Antivirais/administração & dosagem , Antivirais/administração & dosagem , Palivizumab/administração & dosagem , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sinciciais Respiratórios/imunologia , Proteínas Virais de Fusão/imunologia , Animais , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Antivirais/isolamento & purificação , Linhagem Celular , Modelos Animais de Doenças , Humanos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Ligação Proteica , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Resultado do Tratamento
2.
Biometals ; 31(1): 81-89, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29209895

RESUMO

Here we report on the results obtained from an antiviral screening, including herpes simplex virus, vaccinia virus, vesicular stomatitis virus, Coxsackie B4 virus or respiratory syncytial virus, parainfluenza-3 virus, reovirus-1 and Punta Toro virus, of three 2-hydroxy-3-methoxyphenyl acylhydrazone compounds in three cell lines (i.e. human embryonic lung fibroblast cells, human cervix carcinoma cells, and African Green monkey kidney cells). Interesting antiviral EC50 values are obtained against herpes simplex virus-1 and vaccinia virus. The biological activity of acylhydrazones is often attributed to their metal coordinating abilities, so potentiometric and microcalorimetric studies are here discussed to unravel the behavior of the three 2-hydroxy-3-methoxyphenyl compounds in solution. It is worth of note that the acylhydrazone with the higher affinity for Cu(II) ions shows the best antiviral activity against herpes simplex and vaccinia virus (EC50 ~ 1.5 µM, minimal cytotoxic concentration = 60 µM, selectivity index = 40).


Assuntos
Antivirais/farmacologia , Quelantes/farmacologia , Hidrazonas/farmacologia , Simplexvirus/efeitos dos fármacos , Vaccinia virus/efeitos dos fármacos , Animais , Antivirais/síntese química , Antivirais/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Quelantes/síntese química , Quelantes/metabolismo , Chlorocebus aethiops , Cobre/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/virologia , Humanos , Hidrazonas/síntese química , Hidrazonas/metabolismo , Concentração Inibidora 50 , Magnésio/metabolismo , Manganês/metabolismo , Orthoreovirus de Mamíferos/efeitos dos fármacos , Orthoreovirus de Mamíferos/crescimento & desenvolvimento , Orthoreovirus de Mamíferos/metabolismo , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Vírus da Parainfluenza 3 Humana/crescimento & desenvolvimento , Vírus da Parainfluenza 3 Humana/metabolismo , Phlebovirus/efeitos dos fármacos , Phlebovirus/crescimento & desenvolvimento , Phlebovirus/metabolismo , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Vírus Sinciciais Respiratórios/metabolismo , Simplexvirus/crescimento & desenvolvimento , Simplexvirus/metabolismo , Vaccinia virus/crescimento & desenvolvimento , Vaccinia virus/metabolismo , Células Vero , Vesiculovirus/efeitos dos fármacos , Vesiculovirus/crescimento & desenvolvimento , Vesiculovirus/metabolismo
3.
Clin Infect Dis ; 64(suppl_3): S337-S346, 2017 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-28575373

RESUMO

BACKGROUND.: The etiologic inference of identifying a pathogen in the upper respiratory tract (URT) of children with pneumonia is unclear. To determine if viral load could provide evidence of causality of pneumonia, we compared viral load in the URT of children with World Health Organization-defined severe and very severe pneumonia and age-matched community controls. METHODS.: In the 9 developing country sites, nasopharyngeal/oropharyngeal swabs from children with and without pneumonia were tested using quantitative real-time polymerase chain reaction for 17 viruses. The association of viral load with case status was evaluated using logistic regression. Receiver operating characteristic (ROC) curves were constructed to determine optimal discriminatory viral load cutoffs. Viral load density distributions were plotted. RESULTS.: The mean viral load was higher in cases than controls for 7 viruses. However, there was substantial overlap in viral load distribution of cases and controls for all viruses. ROC curves to determine the optimal viral load cutoff produced an area under the curve of <0.80 for all viruses, suggesting poor to fair discrimination between cases and controls. Fatal and very severe pneumonia cases did not have higher viral load than less severe cases for most viruses. CONCLUSIONS.: Although we found higher viral loads among pneumonia cases than controls for some viruses, the utility in using viral load of URT specimens to define viral pneumonia was equivocal. Our analysis was limited by lack of a gold standard for viral pneumonia.


Assuntos
Pneumonia Viral/diagnóstico , Pneumonia Viral/virologia , Infecções Respiratórias/virologia , Carga Viral , Estudos de Casos e Controles , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Internacionalidade , Modelos Logísticos , Masculino , Nasofaringe/virologia , Orofaringe/virologia , Pneumonia Viral/diagnóstico por imagem , Curva ROC , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Vírus Sinciciais Respiratórios/isolamento & purificação , Infecções Respiratórias/microbiologia , Vírus/crescimento & desenvolvimento , Vírus/isolamento & purificação , Organização Mundial da Saúde
4.
Biochem Biophys Res Commun ; 489(4): 460-465, 2017 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-28576489

RESUMO

Respiratory syncytial virus (RSV) is a major cause of respiratory infections in infants and the elderly, leading to more deaths than influenza each year, but there is no antiviral or efficacious vaccine currently available. Here we examine the role in infection of the host mitochondrial protein p32 (HABP/gC1qR/C1qbp) for the first time. RSV replication as well as infectious virus production was significantly reduced by p32 siRNA knockdown, consistent with an important role for p32 in RSV infection. p32 showed distinct mitochondrial localization throughout RSV infection, but immunostaining and high resolution confocal imaging for p32 as well as MitoTracker Red and cytochrome c, revealed clear changes in mitochondrial organization in RSV infection, with perinuclear mitochondrial compaction and asymmetric distribution at 8 and 18 h post-infection, respectively. The results implicate p32 as a key host factor for RSV virus production, and bring to light the potential importance of mitochondria in RSV infection.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas Mitocondriais/metabolismo , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Vírus Sinciciais Respiratórios/metabolismo , Células A549 , Proteínas de Transporte/genética , Humanos , Proteínas Mitocondriais/genética , RNA Interferente Pequeno/genética , Células Tumorais Cultivadas
5.
Pak J Pharm Sci ; 30(4): 1251-1256, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29039322

RESUMO

Isolated Salidroside from the leaves of Nigerian mistletoe (Loranthus micranthus Linn) parasitic on Hevea brasiliensis was evaluated for its antiviral activity against respiratory syncytial virus. Semi- preparative HPLC separation of the ethyl acetate fraction of the leave extract of Loranthus micranthus Linn parasitic on Hevea brasiliensis led to the isolation of a polyphenol. Using spectroscopic methods (1D and 2D NMR and mass spectroscopic data) as well as by comparison with literature data the structure of the compound was determined as 6-O-galloyl salidroside. The antiviral activity of the isolated compound was evaluated against the respiratory syncytial virus. The isolated Salidroside showed potent inhibition towards a recombinant straining respiratory syncytial virus with Inhibitory Concentration (IC 50) value of 10.3±1.50 µg/mL. The result indicates that Salidroside is an efficient antiviral agent against RSV infection and might be useful for the management of RSV pathogenesis.


Assuntos
Antivirais/isolamento & purificação , Antivirais/farmacologia , Hevea/parasitologia , Erva-de-Passarinho/química , Polifenóis/isolamento & purificação , Polifenóis/farmacologia , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Erva-de-Passarinho/crescimento & desenvolvimento , Extratos Vegetais/química , Folhas de Planta/química , Ensaio de Placa Viral
6.
Biochem Biophys Res Commun ; 477(3): 311-6, 2016 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-27346133

RESUMO

Respiratory syncytial virus (RSV) and influenza A virus are leading causes of acute lower respiratory infectious disease. Respiratory diseases caused by RSV and influenza A virus result in serious economic burden and life-threatening disease for immunocompromised people. With the revelation that p38 mitogen-activated protein kinase (MAPK) activity in host cells is crucial for infection and replication of RSV and influenza A virus, inhibition of p38 MAPK activity has been suggested as a potential antiviral therapeutic strategy. However, the low selectivity and high toxicity of the p38 MAPK inhibitors necessitate the development of better inhibitors. Herein, we report the synthesis of a novel p38 MAPK inhibitor, NJK14047, with high kinase selectivity. In this work, it was demonstrated that NJK14047 inhibits RSV- and influenza A-mediated p38 MAPK activation in epithelial cells. Subsequently, NJK14047 treatment resulted in decreased viral replication and viral mRNA synthesis. In addition, secretion of interleukin-6 from infected cells was greatly diminished by NJK14047, suggesting that it can ameliorate immunopathological responses to RSV and influenza A. Collectively, the results suggest that NJK14047 has therapeutic potential to treat respiratory viral infection through the suppression of p38 MAPK activation, which is suggested to be an essential step for respiratory virus infection.


Assuntos
Antivirais/farmacologia , Vírus da Influenza A/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Linhagem Celular , Ativação Enzimática , Vírus da Influenza A/crescimento & desenvolvimento , Vírus da Influenza A/fisiologia , Camundongos , RNA Viral/antagonistas & inibidores , RNA Viral/biossíntese , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Vírus Sinciciais Respiratórios/fisiologia , Ensaio de Placa Viral , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
7.
J Virol ; 89(17): 8974-81, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26085154

RESUMO

UNLABELLED: The small hydrophobic (SH) gene of respiratory syncytial virus (RSV), a major cause of infant hospitalization, encodes a viroporin of unknown function. SH gene knockout virus (RSV ΔSH) is partially attenuated in vivo, but not in vitro, suggesting that the SH protein may have an immunomodulatory role. RSV ΔSH has been tested as a live attenuated vaccine in humans and cattle, and here we demonstrate that it protected against viral rechallenge in mice. We compared the immune response to infection with RSV wild type and RSV ΔSH in vivo using BALB/c mice and in vitro using epithelial cells, neutrophils, and macrophages. Strikingly, the interleukin-1ß (IL-1ß) response to RSV ΔSH infection was greater than to wild-type RSV, in spite of a decreased viral load, and when IL-1ß was blocked in vivo, the viral load returned to wild-type levels. A significantly greater IL-1ß response to RSV ΔSH was also detected in vitro, with higher-magnitude responses in neutrophils and macrophages than in epithelial cells. Depleting macrophages (with clodronate liposome) and neutrophils (with anti-Ly6G/1A8) demonstrated the contribution of these cells to the IL-1ß response in vivo, the first demonstration of neutrophilic IL-1ß production in response to viral lung infection. In this study, we describe an increased IL-1ß response to RSV ΔSH, which may explain the attenuation in vivo and supports targeting the SH gene in live attenuated vaccines. IMPORTANCE: There is a pressing need for a vaccine for respiratory syncytial virus (RSV). A number of live attenuated RSV vaccine strains have been developed in which the small hydrophobic (SH) gene has been deleted, even though the function of the SH protein is unknown. The structure of the SH protein has recently been solved, showing it is a pore-forming protein (viroporin). Here, we demonstrate that the IL-1ß response to RSV ΔSH is greater in spite of a lower viral load, which contributes to the attenuation in vivo. This potentially suggests a novel method by which viruses can evade the host response. As all Pneumovirinae and some Paramyxovirinae carry similar SH genes, this new understanding may also enable the development of live attenuated vaccines for both RSV and other members of the Paramyxoviridae.


Assuntos
Interleucina-1beta/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/imunologia , Proteínas Oncogênicas de Retroviridae/genética , Animais , Linhagem Celular , Células Epiteliais/imunologia , Células Epiteliais/virologia , Feminino , Deleção de Genes , Técnicas de Inativação de Genes , Humanos , Interleucina-1beta/biossíntese , Macrófagos/imunologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/imunologia , Neutrófilos/virologia , Infecções por Vírus Respiratório Sincicial/virologia , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Vacinação , Vacinas Atenuadas/imunologia , Carga Viral/imunologia
8.
Theor Popul Biol ; 110: 78-85, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27155294

RESUMO

Respiratory syncytial virus (RSV) is the main cause of lower respiratory tract infections in children. Whilst highly seasonal, RSV dynamics can have either one-year (annual) or two-year (biennial) cycles. Furthermore, some countries show a 'delayed biennial' pattern, where the epidemic peak in low incidence years is delayed. We develop a compartmental model for RSV infection, driven by a seasonal forcing function, and conduct parameter space and bifurcation analyses to document parameter ranges that give rise to these different seasonal patterns. The model is sensitive to the birth rate, transmission rate, and seasonality parameters, and can replicate RSV dynamics observed in different countries. The seasonality parameter must exceed a threshold for the model to produce biennial cycles. Intermediate values of the birth rate produce the greatest delay in these biennial cycles, while the model reverts to annual cycles if the duration of immunity is too short. Finally, the existence of period doubling and period halving bifurcations suggests robust model dynamics, in agreement with the known regularity of RSV outbreaks. These findings help explain observed RSV data, such as regular biennial dynamics in Western Australia, and delayed biennial dynamics in Finland. From a public health perspective, our findings provide insight into the drivers of RSV transmission, and a foundation for exploring RSV interventions.


Assuntos
Modelos Estatísticos , Infecções por Vírus Respiratório Sincicial/transmissão , Vírus Sincicial Respiratório Humano/patogenicidade , Criança , Surtos de Doenças , Epidemias , Finlândia/epidemiologia , Humanos , Lactente , Infecções por Vírus Respiratório Sincicial/diagnóstico , Infecções por Vírus Respiratório Sincicial/epidemiologia , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Estações do Ano , Austrália Ocidental/epidemiologia
9.
J Virol ; 88(20): 11899-914, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25100835

RESUMO

The small hydrophobic (SH) protein is a 64-amino-acid polypeptide encoded by the human respiratory syncytial virus (hRSV). SH protein has a single α-helical transmembrane (TM) domain that forms pentameric ion channels. Herein, we report the first inhibitor of the SH protein channel, pyronin B, and we have mapped its binding site to a conserved surface of the RSV SH pentamer, at the C-terminal end of the transmembrane domain. The validity of the SH protein structural model used has been confirmed by using a bicellar membrane-mimicking environment. However, in bicelles the α-helical stretch of the TM domain extends up to His-51, and by comparison with previous models both His-22 and His-51 adopt an interhelical/lumenal orientation relative to the channel pore. Neither His residue was found to be essential for channel activity although His-51 protonation reduced channel activity at low pH, with His-22 adopting a more structural role. The latter results are in contrast with previous patch clamp data showing channel activation at low pH, which could not be reproduced in the present work. Overall, these results establish a solid ground for future drug development targeting this important viroporin. Importance: The human respiratory syncytial virus (hRSV) is responsible for 64 million reported cases of infection and 160,000 deaths each year. Lack of adequate antivirals fuels the search for new targets for treatment. The small hydrophobic (SH) protein is a 64-amino-acid polypeptide encoded by hRSV and other paramyxoviruses, and its absence leads to viral attenuation in vivo and early apoptosis in infected cells. SH protein forms pentameric ion channels that may constitute novel drug targets, but no inhibitor for this channel activity has been reported so far. A small-molecule inhibitor, pyronin B, can reduce SH channel activity, and its likely binding site on the SH protein channel has been identified. Black lipid membrane (BLM) experiments confirm that protonation of both histidine residues reduces stability and channel activity. These results contrast with previous patch clamp data that showed low-pH activation, which we have not been able to reproduce.


Assuntos
Bicamadas Lipídicas , Vírus Sinciciais Respiratórios/metabolismo , Proteínas Virais/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Sítios de Ligação , Chlorocebus aethiops , Clonagem Molecular , Genes Virais , Interações Hidrofóbicas e Hidrofílicas , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Técnicas de Patch-Clamp , Pironina/análogos & derivados , Pironina/farmacologia , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Células Vero , Ensaio de Placa Viral , Proteínas Virais/química , Proteínas Virais/genética
10.
J Med Virol ; 87(10): 1796-805, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26243168

RESUMO

The ability of a non-propagating microbial transport medium to maintain the viability of clinically relevant viruses was compared to a similar commercial medium to establish performance equivalence. Two dilutions of stock of test viruses, namely adenovirus (AdV), cytomegalovirus (CMV), echovirus Type 30 (EV), herpes simplex virus (HSV) types 1 and 2, influenza A, parainfluenza 3 (PIV), respiratory syncytial virus (RSV), and varicella zoster virus (VZV), were spiked into Puritan® Medical Products Company Universal Transport System (UniTranz-RT™) and BD(TM) Universal Viral Transport System (UVT) and incubated at 4 °C and room temperature (RT) for up to 72 hr. Post incubation assessment of recovery of AdV, EV, HSV-2, PIV, and VZV from UniTranz-RT™ and UVT using shell vial assays followed by immunofluorescence staining demonstrated statistically significant differences between both transport media. In general, significantly higher recoveries of AdV, EV, and VZV were found from UniTranz-RT™ than UVT whereas HSV-2 and PIV were recovered better from UVT than UniTranz-RT™, under specific test conditions. The recovery of HSV-1, influenza A, PIV, and RSV showed no significant differences between transport media. Sulforhodamine B-based assay analysis of UniTranz-RT™ lots prior to and at expiration exhibited no cytotoxicity. The overall results of the study validate the full performance of UniTranz-RT™ as a viral transport medium and establish its effectiveness on par with the UVT.


Assuntos
Viabilidade Microbiana , Manejo de Espécimes/métodos , Meios de Transporte/métodos , Adenoviridae/crescimento & desenvolvimento , Meios de Cultura , Citomegalovirus/crescimento & desenvolvimento , Herpesvirus Humano 1/crescimento & desenvolvimento , Herpesvirus Humano 2/crescimento & desenvolvimento , Herpesvirus Humano 3/crescimento & desenvolvimento , Humanos , Preservação Biológica/métodos , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Meios de Transporte/normas , Vírus/crescimento & desenvolvimento
11.
Clin Dev Immunol ; 2013: 359683, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23840240

RESUMO

Monoclonal antibodies are widely used both in infants and in adults for several indications. Humanized monoclonal antibodies (palivizumab) have been used for many years for the prevention of respiratory syncytial virus infection in pediatric populations (preterm infants, infants with chronic lung disease or congenital heart disease) at high risk of severe and potentially lethal course of the infection. This drug was reported to be safe, well tolerated and effective to decrease the hospitalization rate and mortality in these groups of infants by several clinical trials. In the present paper we report the development and the current use of monoclonal antibodies for prophylaxis against respiratory syncytial virus.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Antivirais/uso terapêutico , Cardiopatias Congênitas/tratamento farmacológico , Doenças do Prematuro/prevenção & controle , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Adulto , Anticorpos Monoclonais Humanizados/farmacologia , Antivirais/farmacologia , Pré-Escolar , Ensaios Clínicos como Assunto , Cardiopatias Congênitas/imunologia , Cardiopatias Congênitas/mortalidade , Cardiopatias Congênitas/virologia , Hospitalização , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/imunologia , Doenças do Prematuro/mortalidade , Doenças do Prematuro/virologia , Palivizumab , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/mortalidade , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Análise de Sobrevida
12.
Antimicrob Agents Chemother ; 56(9): 4927-36, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22802243

RESUMO

Although it has been on the market for over a decade, confusion remains regarding the pharmacokinetics (PK) and optimal dosing of palivizumab, a humanized IgG1κ monoclonal antibody indicated for the prevention of serious lower respiratory tract disease caused by respiratory syncytial virus (RSV) in pediatric patients at high risk of RSV disease. The objectives of this analysis were to characterize the population PK of palivizumab in adults and children using nonlinear mixed-effect modeling, quantify the effects of individual covariates on variability in palivizumab disposition, and compare palivizumab exposures for various dosing scenarios. Palivizumab PK data from 22 clinical studies were used for model development. The model was developed using a two-stage approach: (i) a 2-compartment model with first-order absorption after intramuscular administration was fitted to adult data, and (ii) the same structural model was fitted to the sparse pediatric data using the NONMEM $PRIOR subroutine, with informative priors obtained from the adult analysis. Body weight and an age descriptor that combines gestational age and postnatal age (PAGE) using an asymptotic-exponential model best described palivizumab clearance in pediatric patients. Palivizumab clearance increased slightly from 10.2 ml/day to 11.9 ml/day as a function of PAGE ranging from 7 to 18 months. Covariate analysis indicated a 20% higher clearance in children with chronic lung disease and in children with antidrug antibody titer values of ≥80. These covariates did not substantially explain interindividual variability. In the label-indicated pediatric population, body weight was the primary demographic factor affecting palivizumab PK. Body weight-based dosing of 15 mg/kg yields similar palivizumab concentrations in children of different gestational and postnatal ages. Simulations demonstrated that there was little difference in palivizumab PK between healthy term and premature infants. Simulations also demonstrated that the 5 monthly palivizumab doses of 15 mg/kg, consistent with the label and studied in two randomized, clinical trials, provided greater and more prolonged palivizumab exposure than did an abbreviated dosing regimen of 3 monthly doses.


Assuntos
Anticorpos Monoclonais Humanizados , Antivirais , Doenças do Prematuro/tratamento farmacológico , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Adulto , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/farmacocinética , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Antivirais/administração & dosagem , Antivirais/farmacocinética , Peso Corporal , Criança , Vias de Administração de Medicamentos , Esquema de Medicação , Feminino , Idade Gestacional , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/virologia , Masculino , Modelos Estatísticos , Palivizumab , Ensaios Clínicos Controlados Aleatórios como Assunto , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento
13.
J Gen Virol ; 93(Pt 3): 565-576, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22170635

RESUMO

The cytidine deaminase APOBEC3G (apolipoprotein B mRNA-editing enzyme-catalytic polypeptide 3G; A3G) exerts antiviral activity against retroviruses, hepatitis B virus, adeno-associated virus and transposable elements. We assessed whether the negative-strand RNA viruses measles, mumps and respiratory syncytial might be affected by A3G, and found that their infectivity was reduced by 1-2 logs (90-99 %) in A3G overexpressing Vero cells, and in T-cell lines expressing A3G at physiological levels. Viral RNA was co-precipitated with HA-tagged A3G and could be amplified by RT-PCR. Interestingly, A3G reduced viral transcription and protein expression in infected cells by 50-70 %, and caused an increased mutation frequency of 0.95 mutations per 1000 nt in comparison to the background level of 0.22/1000. The observed mutations were not specific for A3G [cytidine to uridine (C→U) or guanine to adenine (G→A) hypermutations], nor specific for ADAR (adenosine deaminase acting on RNA, A→G and U→C transitions, with preference for next neighbour-nucleotides U = A>C>G). In addition, A3G mutants with inactivated catalytic deaminase (H257R and E259Q) were inhibitory, indicating that the deaminase activity is not required for the observed antiviral activity. In combination, impaired transcription and increased mutation frequencies are sufficient to cause the observed reduction in viral infectivity and eliminate virus replication within a few passages in A3G-expressing cells.


Assuntos
Citidina Desaminase/metabolismo , Vírus do Sarampo/patogenicidade , Vírus da Caxumba/patogenicidade , Vírus Sinciciais Respiratórios/patogenicidade , Replicação Viral , Desaminase APOBEC-3G , Animais , Antivirais/metabolismo , Linhagem Celular , Citidina Desaminase/imunologia , Humanos , Vírus do Sarampo/crescimento & desenvolvimento , Vírus do Sarampo/imunologia , Vírus da Caxumba/crescimento & desenvolvimento , Vírus da Caxumba/imunologia , Mutação Puntual , RNA Viral/genética , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Vírus Sinciciais Respiratórios/imunologia
14.
J Virol ; 85(10): 4792-801, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21389127

RESUMO

Respiratory syncytial virus (RSV) infection of most cultured cell lines causes cell-cell fusion and death. Cell fusion is caused by the fusion (F) glycoprotein and is clearly cytopathic, but other aspects of RSV infection may also contribute to cytopathology. To investigate this possibility, we generated an RSV replicon that lacks all three of its glycoprotein genes and so cannot cause cell-cell fusion or virus spread. This replicon includes a green fluorescent protein gene and an antibiotic resistance gene to enable detection and selection of replicon-containing cells. Adaptive mutations in the RSV replicon were not required for replicon maintenance. Cells containing the replicon could be cloned and passaged many times in the absence of antibiotic selection, with 99% or more of the cells retaining the replicon after each cell division. Transient expression of the F and G (attachment) glycoproteins supported the production of virions that could transfer the replicon into most cell lines tested. Since the RSV replicon is not toxic to these cultured cells and does not affect their rate of cell division, none of the 8 internal viral proteins, the viral RNA transcripts, or the host response to these molecules or their activities is cytopathic. However, the level of replicon genome and gene expression is controlled in some manner well below that of complete virus and, as such, might avoid cytotoxicity. RSV replicons could be useful for cytoplasmic gene expression in vitro and in vivo and for screening for compounds active against the viral polymerase.


Assuntos
Expressão Gênica , Proteínas Recombinantes/biossíntese , Replicon , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Vírus Sinciciais Respiratórios/genética , Deleção de Genes , Genes Virais/genética , Vetores Genéticos , Instabilidade Genômica , Proteínas Recombinantes/genética , Proteínas Estruturais Virais/genética
15.
Am J Respir Crit Care Med ; 182(10): 1305-14, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20622030

RESUMO

RATIONALE: Respiratory syncytial virus (RSV) is the leading cause of childhood lower respiratory infection, yet viable therapies are lacking. Two major challenges have stalled antiviral development: ethical difficulties in performing pediatric proof-of-concept studies and the prevailing concept that the disease is immune-mediated rather than being driven by viral load. OBJECTIVES: The development of a human experimental wild-type RSV infection model to address these challenges. METHODS: Healthy volunteers (n = 35), in five cohorts, received increasing quantities (3.0-5.4 log plaque-forming units/person) of wild-type RSV-A intranasally. MEASUREMENTS AND MAIN RESULTS: Overall, 77% of volunteers consistently shed virus. Infection rate, viral loads, disease severity, and safety were similar between cohorts and were unrelated to quantity of RSV received. Symptoms began near the time of initial viral detection, peaked in severity near when viral load peaked, and subsided as viral loads (measured by real-time polymerase chain reaction) slowly declined. Viral loads correlated significantly with intranasal proinflammatory cytokine concentrations (IL-6 and IL-8). Increased viral load correlated consistently with increases in multiple different disease measurements (symptoms, physical examination, and amount of nasal mucus). CONCLUSIONS: Viral load appears to drive disease manifestations in humans with RSV infection. The observed parallel viral and disease kinetics support a potential clinical benefit of RSV antivirals. This reproducible model facilitates the development of future RSV therapeutics.


Assuntos
Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/patogenicidade , Carga Viral , Adolescente , Adulto , Quimiocinas/análise , Citocinas/análise , Feminino , Humanos , Interleucina-6/análise , Masculino , Pessoa de Meia-Idade , Líquido da Lavagem Nasal/imunologia , Infecções por Vírus Respiratório Sincicial/etiologia , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Ensaio de Placa Viral , Adulto Jovem
16.
J Virol Methods ; 293: 114120, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33676967

RESUMO

BACKGROUND: Primary rhesus monkey kidney cells (RhMK) can be used for the detection of respiratory viruses, including influenza and parainfluenza. The human colon adeno-carcinoma cell line, CACO-2, has been previously used for the growth of multiple influenza viruses, including seasonal, novel and avian lineages. OBJECTIVE: We compared CACO-2, Madin-Darby Canine Kidney (MDCK), and RhMK cells for the isolation of viruses from patients presenting with influenza like-illness (ILI). STUDY DESIGN: Nasopharyngeal specimens from patients with ILI in primary care settings were processed for conventional viral culture in MDCK, RhMK, and CACO-2. Cells were examined microscopically for cytopathic effect (CPE) and confirmatory testing included immunofluorescent antigen (IFA) detection and real-time RT-PCR. Additionally, 16 specimens positive for respiratory syncytial virus (RSV) by PCR were inoculated on CACO-2 cells. Statistical analysis was done using Chi-square test with IBM Statistical Program. RESULTS: Of 1031 respiratory specimens inoculated, viruses were isolated and confirmed from 331 (32.1 %) in MDCK cells, 304 (29.5 %) in RhMk cells, and 433 (42.0 %) in CACO-2 cells. These included influenza A/(H1N1)pdm09, influenza A(H3N2), influenza B, parainfluenza virus (PIV) types 1, 2, and 3, human coronavirus 229E (CoV-229E), human adenovirus (HAdV), herpes simplex virus 1 (HSV 1), and enterovirus (EV). Influenza A viruses grew best in the CACO-2 cell line. Time to observation of CPE was similar for all three cell types but unlike RhMK and MDCK cells, virus-specific morphological changes were indistinguishable in CACO-2 cells. None of the 16 specimens positive for RSV by PCR grew on CACO-2 cells. CONCLUSIONS: The overall respiratory virus culture isolation rate in CACO-2 cells was significantly higher than that in RhMK or MDCK cells (p < 0.05). CACO-2 cells also supported the growth of some viruses that did not grow in either RhMK or MDCK cells. Except for RSV, CACO-2 cells provide a worthwhile addition to culture algorithms for respiratory specimens.


Assuntos
Influenza Humana/virologia , Nasofaringe/virologia , Adenovírus Humanos/crescimento & desenvolvimento , Adenovírus Humanos/isolamento & purificação , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Células CACO-2 , Criança , Pré-Escolar , Cães , Feminino , Humanos , Lactente , Células Madin Darby de Rim Canino , Masculino , Pessoa de Meia-Idade , Orthomyxoviridae/crescimento & desenvolvimento , Orthomyxoviridae/isolamento & purificação , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Vírus Sinciciais Respiratórios/isolamento & purificação , Adulto Jovem
17.
Biochem Pharmacol ; 177: 113982, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32305436

RESUMO

Marine environments are known to be a new source of structurally diverse bioactive molecules. In this paper, we identified a porphyrin derivative of Pyropheophorbide a (PPa) from the mussel Musculus senhousei (M. senhousei) that showed broad anti-influenza A virus activity in vitro against a panel of influenza A viral strains. The analysis of the mechanism of action indicated that PPa functions in the early stage of virus infection by interacting with the lipid bilayer of the virion, resulting in an alteration of membrane-associated functions, thereby blocking the entry of enveloped viruses into host cells. In addition, the anti-influenza A virus activity of PPa was further assessed in mice infected with the influenza A virus. The survival rate and mean survival time of mice were apparently prolonged compared with the control group which was not treated with the drug. Therefore, PPa and its derivatives may represent lead compounds for controlling influenza A virus infection.


Assuntos
Antivirais/farmacologia , Betacoronavirus/efeitos dos fármacos , Bivalves/química , Clorofila/análogos & derivados , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Vírion/efeitos dos fármacos , Animais , Antivirais/química , Antivirais/isolamento & purificação , Betacoronavirus/crescimento & desenvolvimento , Betacoronavirus/metabolismo , Clorofila/química , Clorofila/isolamento & purificação , Clorofila/farmacologia , Cães , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H1N1/metabolismo , Bicamadas Lipídicas/antagonistas & inibidores , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Células Madin Darby de Rim Canino , Masculino , Camundongos , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/mortalidade , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Vírus Sinciciais Respiratórios/metabolismo , SARS-CoV-2 , Alimentos Marinhos , Análise de Sobrevida , Vírion/crescimento & desenvolvimento , Vírion/metabolismo , Internalização do Vírus/efeitos dos fármacos
18.
Mikrobiyol Bul ; 43(3): 433-8, 2009 Jul.
Artigo em Turco | MEDLINE | ID: mdl-19795618

RESUMO

Respiratory Syncytial Virus (RSV) is the most important viral agent leading to lower respiratory tract infection in infants and children. The aim of this study was to investigate the presence of RSV by direct immunofluorescence antibody (DFA), cell culture and polymerase chain reaction (PCR) in children with lower respiratory tract infection. Nasotracheal aspirate specimens collected from 80 hospitalized patients aged between 0-24 months and clinically diagnosed as lower respiratory tract infection, during November 2005-May 2006 period, were included to the study. RSV antigen was investigated in clinical specimens by DFA method (Monofluo Bio-Rad, France). Hep-2 culture was used for isolation of RSV. RSV-RNA was investigated by real-time PCR (Fluorion lontek, Turkey) in clinical specimens. RSV was found positive in 26 (32.5%) of 80 samples by DFA and in 17 (21.3%) samples by cell culture. Six specimens were not studied by PCR as sample amounts were not sufficient. Of the 74 samples tested, 20 (27%) were found to be positive by real-time PCR. Fifty-four of the samples were negative by 3 of the methods, while 12 were positive by all of them. DFA and PCR positive 8 samples yielded negative result in cell culture. Five of the 6 samples not investigated by PCR, were positive both in DFA and cell culture while 1 sample was positive only by DFA. Considering cell culture as the gold standard, the sensitivity, specificity, positive and negative predictive values were found as 100%, 85.7%, 65.4% and 100%, respectively, for DFA and 100%, 94.7%, 85% and 100%, respectively, for PCR. As a conclusion for the accurate diagnosis of RSV infections the clinical samples should be collected in the early phase of the disease and inoculated to the cell cultures immediately for viral isolation. If cell culture or PCR facilities are not available for routine diagnosis, DFA method can be used for rapid and cost effective diagnosis of RSV infections.


Assuntos
Técnica Direta de Fluorescência para Anticorpo/normas , Reação em Cadeia da Polimerase/normas , Infecções por Vírus Respiratório Sincicial/diagnóstico , Vírus Sinciciais Respiratórios/isolamento & purificação , Infecções Respiratórias/diagnóstico , Antígenos Virais/análise , Linhagem Celular , Pré-Escolar , Humanos , Lactente , Recém-Nascido , Mucosa Nasal/virologia , Valor Preditivo dos Testes , RNA Viral/análise , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Vírus Sinciciais Respiratórios/imunologia , Infecções Respiratórias/virologia , Sensibilidade e Especificidade , Traqueia/virologia
19.
Elife ; 82019 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-31246170

RESUMO

Although respiratory syncytial virus (RSV) is responsible for more human deaths each year than influenza, its pathogenic mechanisms are poorly understood. Here high-resolution quantitative imaging, bioenergetics measurements and mitochondrial membrane potential- and redox-sensitive dyes are used to define RSV's impact on host mitochondria for the first time, delineating RSV-induced microtubule/dynein-dependent mitochondrial perinuclear clustering, and translocation towards the microtubule-organizing centre. These changes are concomitant with impaired mitochondrial respiration, loss of mitochondrial membrane potential and increased production of mitochondrial reactive oxygen species (ROS). Strikingly, agents that target microtubule integrity the dynein motor protein, or inhibit mitochondrial ROS production strongly suppresses RSV virus production, including in a mouse model with concomitantly reduced virus-induced lung inflammation. The results establish RSV's unique ability to co-opt host cell mitochondria to facilitate viral infection, revealing the RSV-mitochondrial interface for the first time as a viable target for therapeutic intervention.


Assuntos
Interações Hospedeiro-Patógeno , Mitocôndrias/patologia , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Replicação Viral , Células A549 , Animais , Modelos Animais de Doenças , Dineínas/metabolismo , Humanos , Pulmão/patologia , Pulmão/virologia , Camundongos , Microtúbulos/metabolismo , Infecções por Vírus Respiratório Sincicial/patologia
20.
J Virol Methods ; 263: 88-95, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30381239

RESUMO

Viral plaque assays are important tools in the development and evaluation of new antiviral drugs or vaccines in both preclinical and clinical research. While plaque assays are the standard tools to measure infectious virus, the methodology is time-consuming and requires experience in recognizing plaques. The assays are also prone to variation among analysts due to plaque recognition and manual counting errors. Here we describe the development of two simplified plaque assays for measuring RSV virus titers and anti-RSV antibody neutralization titers using 96 well plate formats. First, we evaluated multiple parameters to build up a quantitative plaque assay to measure infectious RSV. We then optimized the assay conditions to assess the fundamental changes from the traditional plaque assay, which were elimination of overnight pre-seeding host cells and addition of a centrifugation step after viral infection of the cells. We designed DoE to refine four key parameters within one experiment for host cell density, host cell volume, viral inoculum volume, host cell and viral mixture incubation time to make this assay more robust. We have also adapted these conditions into a second assay, which was an automated plaque reduction neutralization assay (PRNT) to determine neutralization titers of anti-RSV antibodies. Both assays utilize immune fluorescence staining to detect viral plaques. The images of the immuno-stained wells are captured by the PerkinElmer EnSight instrument and show clear visualization of plaques harvesting on day 3. Software algorithm was specifically designed for automatic counting of these fluorescent "objects". The quantitative plaque assay provided titers of RSV similar to those obtained from the traditional plaque assay. The method has been successfully utilized to screen multiple vaccine candidates in viral shedding efficacy studies. The automated PRNT assay provided antibody neutralizing titers that matched with published data. This automated 96 well plaque assay has made it possible to screen RSV samples in a higher throughput manner, and can be extended to other infectious organisms that form plaques for vaccine or drug evaluation.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Imagem Óptica , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/crescimento & desenvolvimento , Ensaio de Placa Viral/métodos , Algoritmos , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Linhagem Celular Tumoral , Modelos Animais de Doenças , Avaliação de Medicamentos , Feminino , Humanos , Testes de Neutralização , Reprodutibilidade dos Testes , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Sigmodontinae/imunologia , Sigmodontinae/virologia , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA