Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Lancet Glob Health ; 7(7): e940-e950, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31200893

RESUMO

BACKGROUND: Paediatric vaccination against influenza can result in indirect protection, by reducing transmission to their unvaccinated contacts. We investigated whether influenza vaccination of children would protect them and their household members in a resource-limited setting. METHODS: We did a cluster-randomised, blinded, controlled study in three villages in India. Clusters were defined as households (ie, dwellings that shared a courtyard), and children aged 6 months to 10 years were eligible for vaccination as and when they became age-eligible throughout the study. Households were randomly assigned (1:1) by a computer-based system to intramuscular trivalent inactivated influenza vaccine (IIV3) or a control of inactivated poliovirus vaccine (IPV) in the beginning of the study; vaccination occurred once a year for 3 years. The primary efficacy outcome was laboratory-confirmed influenza in a vaccinated child with febrile acute respiratory illness, analysed in the modified intention-to-treat population (ie, children who received at least one dose of vaccine, were under surveillance, and had not an influenza infection within 15 days of last vaccine dose). The secondary outcome for indirect effectiveness (surveillance study) was febrile acute respiratory illness in an unvaccinated household member of a vaccine study participant. Data from each year (year 1: November, 2009, to October, 2010; year 2: October, 2010, to October, 2011; and year 3: October, 2011, to May, 2012) were analysed separately. Safety was analysed among all participants who were vaccinated with at least one dose of the vaccine. This trial is registered with ClinicalTrials.gov, number NCT00934245. FINDINGS: Between Nov 1, 2009, to May 1, 2012, we enrolled 3208 households, of which 1959 had vaccine-eligible children. 1010 households were assigned to IIV3 and 949 households were assigned to IPV. In 3 years, we vaccinated 4345 children (2132 with IIV3 and 2213 with IPV) from 1868 households (968 with IIV3 and 900 with IPV) with 10 813 unvaccinated household contacts. In year 1, influenza virus was detected in 151 (10%) of 1572 IIV3 recipients and 206 (13%) of 1633 of IPV recipients (total IIV3 vaccine efficacy 25·6% [95% CI 6·8-40·6]; p=0·010). In year 2, 105 (6%) of 1705 IIV3 recipients and 182 (10%) of 1814 IPV recipients had influenza (vaccine efficacy 41·0% [24·1-54·1]; p<0·0001). In year 3, 20 (1%) of 1670 IIV3 recipients and 81 (5%) of 1786 IPV recipients had influenza (vaccine efficacy 74·2% [57·8-84·3]; p<0·0001). In year 1, total vaccine efficacy against influenza A(H1N1)pdm09 was 14·5% (-20·4 to 39·3). In year 2, total vaccine efficacy against influenza A(H3N2) was 64·5% (48·5-75·5). Total vaccine efficacy against influenza B was 32·5% (11·3-48·6) in year 1, 4·9% (-38·9 to 34·9) in year 2, and 76·5% (59·4-86·4) in year 3. Indirect vaccine effectiveness was statistically significant only in year 3 (38·1% [7·4-58·6], p=0·0197) when influenza was detected in 39 (1%) of 4323 IIV3-allocated and 60 (1%) of 4121 IPV-allocated household unvaccinated individuals. In the IIV3 group, 225 (12%) of 1632 children in year 1, 375 (22%) of 1718 in year 2, and 209 (12%) of 1673 in year 3 had an adverse reaction (compared with 216 [13%] of 1730, 380 [21%] of 1825, and 235 [13%] of 1796, respectively, in the IPV group). The most common reactions in both groups were fever and tenderness at site. No vaccine-related deaths occurred in either group. INTERPRETATION: IIV3 provided variable direct and indirect protection against influenza infection. Indirect protection was significant during the year of highest direct protection and should be considered when quantifying the effect of vaccination programmes. FUNDING: US Centers for Disease Control and Prevention.


Assuntos
Vacinas contra Influenza/administração & dosagem , Influenza Humana/prevenção & controle , População Rural , Vacinas de Produtos Inativados/administração & dosagem , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Análise por Conglomerados , Feminino , Humanos , Índia , Lactente , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento , Adulto Jovem
2.
J Med Virol ; 83(10): 1799-810, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21837798

RESUMO

Human metapneumovirus (hMPV) causes acute respiratory infections in children and adults. It is classified into two major genetic lineages and each lineage into two sublineages. The purpose of the study was to identify and characterize hMPV in children who presented to the All India Institute of Medical Sciences, New Delhi, India with acute respiratory infection from April 2005 to March 2007. By reverse-transcription polymerase chain reaction, hMPV was detected in 21 (3%) of the 662 nasopharyngeal samples from children with acute respiratory infection and in none of the 120 control children. Seven of the 21 (33%) children infected with hMPV required hospital admission for pneumonia or bronchiolitis. Most hMPV detections were during the winter and spring seasons. The majority (67%, 11/21) of children positive for hMPV were within 24 months of age. Phylogenetic analysis of partial F and N gene and the full G gene sequences showed three sub-lineages of hMPV circulated during the study period, B1, B2, and the novel sub-lineage A2b. The circulation pattern of hMPV genotypes varied by season. Comparison of the F and G genes of eight strains revealed incongruencies in lineage assignments, raising the possibility that recombination had occurred. Sequence analysis also revealed the F gene was relatively conserved whereas the G gene was more variable between the A and B lineages. This study demonstrates that hMPV is an important contributor to acute respiratory infection in children in India, resulting in both outpatient visits and hospitalizations.


Assuntos
Variação Genética , Metapneumovirus/genética , Metapneumovirus/isolamento & purificação , Infecções por Paramyxoviridae/virologia , Infecções Respiratórias/epidemiologia , Infecções Respiratórias/virologia , Sequência de Aminoácidos , Sequência de Bases , Criança , Pré-Escolar , Feminino , Genótipo , Humanos , Índia , Lactente , Masculino , Metapneumovirus/classificação , Nasofaringe/virologia , Infecções por Paramyxoviridae/diagnóstico , Infecções por Paramyxoviridae/epidemiologia , Filogenia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Análise de Sequência de RNA
3.
J Med Virol ; 82(5): 812-6, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20336746

RESUMO

Human bocavirus (HBoV) is a new human parvovirus identified in children with respiratory tract disease. Nasopharyngeal aspirates were collected from 305 children <5 years of age with acute respiratory tract infection from April 2005 to March 2007 and screened for the presence of HBoV by two separate sets of a polymerase chain reaction (PCR) described previously. Twenty-two (7.2%) children who had acute respiratory infection were found to be positive for HBoV by both sets of PCR. The main clinical symptoms were cough (95%), runny nose (64%), and fever (59%). In two samples, HBoV was identified together with respiratory syncytial virus in one sample and influenza A virus in another. HBoV appeared to have no seasonal distribution and is associated with both upper and lower respiratory tract disease in young children in India.


Assuntos
Bocavirus Humano/isolamento & purificação , Infecções por Parvoviridae/epidemiologia , Infecções Respiratórias/epidemiologia , Pré-Escolar , Feminino , Humanos , Índia/epidemiologia , Lactente , Vírus da Influenza A/isolamento & purificação , Masculino , Nasofaringe/virologia , Infecções por Parvoviridae/patologia , Infecções por Parvoviridae/virologia , Reação em Cadeia da Polimerase/métodos , Prevalência , Vírus Sinciciais Respiratórios/isolamento & purificação , Infecções Respiratórias/patologia , Infecções Respiratórias/virologia
4.
Virol J ; 6: 89, 2009 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-19558656

RESUMO

BACKGROUND: Acute lower respiratory tract infections (ALRI) are the major cause of morbidity and mortality in young children worldwide. Information on viral etiology in ALRI from India is limited. The aim of the present study was to develop a simple, sensitive, specific and cost effective multiplex PCR (mPCR) assay without post PCR hybridization or nested PCR steps for the detection of respiratory syncytial virus (RSV), influenza viruses, parainfluenza viruses (PIV1-3) and human metapneumovirus (hMPV). Nasopharyngeal aspirates (NPAs) were collected from children with ALRI < or = 5 years of age. The sensitivity and specificity of mPCR was compared to virus isolation by centrifugation enhanced culture (CEC) followed by indirect immunofluorescence (IIF). RESULTS: From April 2005-March 2007, 301 NPAs were collected from children attending the outpatient department or admitted to the ward of All India Institute of Medical Sciences hospital at New Delhi, India. Multiplex PCR detected respiratory viruses in 106 (35.2%) of 301 samples with 130 viruses of which RSV was detected in 61, PIV3 in 22, PIV2 in 17, hMPV in 11, PIV1 in 10 and influenza A in 9 children. CEC-IIF detected 79 viruses only. The sensitivity of mPCR was 0.1TCID50 for RSV and influenza A and 1TCID50 for hMPV, PIV1, PIV2, PIV3 and Influenza B. Mixed infections were detected in 18.8% of the children with viral infections, none detected by CEC-IIF. Bronchiolitis was significantly associated with both total viral infections and RSV infection (p < 0.05). History of ARI in family predisposed children to acquire viral infection (p > 0.05). CONCLUSION: Multiplex PCR offers a rapid, sensitive and reasonably priced diagnostic method for common respiratory viruses.


Assuntos
Reação em Cadeia da Polimerase/métodos , Infecções Respiratórias/virologia , Viroses/diagnóstico , Viroses/virologia , Vírus/isolamento & purificação , Pré-Escolar , Hospitais , Humanos , Índia , Lactente , Nasofaringe/virologia , Reação em Cadeia da Polimerase/economia , Sensibilidade e Especificidade , População Urbana , Vírus/genética
5.
J Biol Chem ; 284(11): 7294-306, 2009 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-19131335

RESUMO

Respiratory syncytial virus (RSV) infection has been shown to reduce Na+-driven alveolar fluid clearance in BALB/c mice in vivo. To investigate the cellular mechanisms by which RSV inhibits amiloride-sensitive epithelial Na+ channels (ENaC), the main pathways through which Na+ ions enter lung epithelial cells, we infected human Clara-like lung (H441) cells with RSV that expresses green fluorescent protein (rRA2). 3-6 days later patch clamp recordings showed that infected cells (i.e. cells expressing green fluorescence; GFP+) had significantly lower whole-cell amiloride-sensitive currents and single channel activity (NPo) as compared with non-infected (GFP-), non-inoculated, or cells infected with UV-inactivated RSV. Both alpha and beta ENaC mRNA levels were significantly reduced in GFP+ cells as measured by real-time reverse transcription-PCR. Infection with RSV increased expression of the inducible nitric-oxide synthase (iNOS) and nitrite concentration in the culture medium; nuclear translocation of NF-kappaB p65 subunit and NF-kappaB activation were also up-regulated. iNOS up-regulation in GFP+ cells was prevented by knocking down IkappaB kinase gamma before infection. Furthermore, pretreatment of H441 cells with the specific iNOS inhibitor 1400W (1 microM) resulted in a doubling of the amiloride-sensitive Na+ current in GFP+ cells. Additionally, preincubation of H441 cells with A77-1726 (20 microM), a de novo UTP synthesis inhibitor, and 1400W completely reversed the RSV inhibition of amiloride-sensitive currents in GFP+ cells. Thus, both UTP- and iNOS-generated reactive species contribute to ENaC down-regulation in RSV-infected airway epithelial cells.


Assuntos
Células Epiteliais/metabolismo , Canais Epiteliais de Sódio/biossíntese , Óxido Nítrico Sintase Tipo II/metabolismo , Mucosa Respiratória/metabolismo , Infecções por Vírus Respiratório Sincicial/metabolismo , Vírus Sinciciais Respiratórios/metabolismo , Uridina Trifosfato/metabolismo , Compostos de Anilina/farmacologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Linhagem Celular , Crotonatos , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Células Epiteliais/virologia , Canais Epiteliais de Sódio/genética , Técnicas de Silenciamento de Genes , Humanos , Hidroxibutiratos/farmacologia , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Pulmão , Camundongos , Camundongos Endogâmicos BALB C , Óxido Nítrico Sintase Tipo II/genética , Nitrilas , Nitritos/metabolismo , Mucosa Respiratória/virologia , Infecções por Vírus Respiratório Sincicial/genética , Vírus Sinciciais Respiratórios/genética , Sódio , Toluidinas , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Uridina Trifosfato/genética
6.
Am J Respir Cell Mol Biol ; 40(5): 588-600, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-18952569

RESUMO

We investigated the mechanisms by which respiratory syncytial virus (RSV) infection decreases vectorial Na+ transport across respiratory epithelial cells. Mouse tracheal epithelial (MTE) cells from either BALB/c or C57BL/6 mice and human airway H441 cells were grown on semipermeable supports under an air-liquid interface. Cells were infected with RSV-A2 and mounted in Ussing chambers for measurements of short-circuit currents (I(sc)). Infection with RSV for 24 hours (multiplicity of infection = 1) resulted in positive immunofluorescence for RSV antigen in less than 10% of MTE or H441 cells. In spite of the limited number of cells infected, RSV reduced both basal and amiloride-sensitive I(sc) in both MTE and H441 cells by approximately 50%, without causing a concomitant reduction in transepithelial resistance. Agents that increased intracellular cAMP (forskolin, cpt-CAMP, and IBMX) increased mainly Cl(-) secretion in MTE cells and Na+ absorption in H441 cells. RSV infection for 24 hours blunted both variables. In contrast, ouabain sensitive I(sc), measured across apically permeabilized H441 monolayers, remained unchanged. Western blot analysis of H441 cell lysates demonstrated reductions in alpha- but not gamma-ENaC subunit protein levels at 24 hours after RSV infection. The reduction in amiloride-sensitive I(sc) in H441 cells was prevented by pretreatment with inhibitors of de novo pyrimidine or purine synthesis (A77-1726 and 6-MP, respectively, 50 microM). Our results suggest that infection of both murine and human respiratory epithelial cells with RSV inhibits vectorial Na+ transport via nucleotide release. These findings are consistent with our previous studies showing reduced alveolar fluid clearance after RSV infection of BALB/c mice.


Assuntos
Células Epiteliais/metabolismo , Células Epiteliais/virologia , Pulmão/citologia , Pulmão/virologia , Infecções por Vírus Respiratório Sincicial/metabolismo , Sódio/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico Ativo/efeitos dos fármacos , Linhagem Celular , Colforsina/farmacologia , Células Epiteliais/efeitos dos fármacos , Canais Epiteliais de Sódio/metabolismo , Imunofluorescência , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Pulmão/metabolismo , Camundongos , Subunidades Proteicas/metabolismo , Purinas/biossíntese , Pirimidinas/biossíntese , Terbutalina/farmacologia , Traqueia/citologia , Traqueia/virologia , Uridina Trifosfato/farmacologia
7.
Am J Respir Cell Mol Biol ; 37(4): 379-86, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17541010

RESUMO

Despite respiratory syncytial virus (RSV) bronchiolitis remaining the most common cause of lower respiratory tract disease in infants worldwide, treatment has progressed little in the past 30 years. The aim of our study was to determine whether post-infection administration of de novo pyrimidine synthesis inhibitors could prevent the reduction in alveolar fluid clearance (AFC) and hypoxemia that occurs at Day 2 after intranasal infection of BALB/c mice with RSV. BALB/c mice were infected intranasally with RSV strain A2. AFC was measured in anesthetized, ventilated mice after instillation of 5% bovine serum albumin into the dependent lung. Post-infection systemic treatment with leflunomide has no effect on AFC. However, when added to the AFC instillate, leflunomide's active metabolite, A77-1726, blocks RSV-mediated inhibition of AFC at Day 2. This block is reversed by uridine (which allows pyrimidine synthesis via the scavenger pathway) and not recapitulated by genistein (which mimics the tyrosine kinase inhibitor effects of A77-1726), indicating that the effect is specific for the de novo pyrimidine synthesis pathway. More importantly, when administered intranasally at Day 1, A77-1726, but not its vehicle dimethyl sulfoxide, maintains its beneficial effect on AFC and lung water content until Day 2. Intranasal instillation of A77-1726 at Day 1 also reduces bronchoalveolar lavage nucleotide levels, lung inflammation, and hypoxemia at Day 2 without impairing viral replication at Day 2 or viral clearance at Day 8. Post-infection intranasal or aerosolized treatment with pyrimidine synthesis inhibitors may provide symptomatic relief from the pathophysiologic sequelae of impaired AFC in children with RSV bronchiolitis.


Assuntos
Compostos de Anilina/uso terapêutico , Antivirais/uso terapêutico , Hidroxibutiratos/uso terapêutico , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Infecções por Vírus Respiratório Sincicial/patologia , Administração Intranasal , Resistência das Vias Respiratórias/efeitos dos fármacos , Compostos de Anilina/administração & dosagem , Compostos de Anilina/farmacologia , Animais , Antivirais/administração & dosagem , Antivirais/farmacologia , Líquido da Lavagem Broncoalveolar/citologia , Contagem de Células , Quimiocinas/metabolismo , Crotonatos , DNA/biossíntese , Dimetil Sulfóxido/farmacologia , Esquema de Medicação , Água Extravascular Pulmonar/efeitos dos fármacos , Água Extravascular Pulmonar/virologia , Hidroxibutiratos/administração & dosagem , Hidroxibutiratos/farmacologia , Isoxazóis/farmacologia , Leflunomida , Pulmão/efeitos dos fármacos , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Nitrilas , Oxigênio/sangue , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Vírus Sinciciais Respiratórios/fisiologia , Toluidinas , Replicação Viral/efeitos dos fármacos
8.
PLoS One ; 2(6): e491, 2007 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-17551572

RESUMO

BACKGROUND: Acute respiratory infection (ARI) is a major killer of children in developing countries. Although the frequency of ARI is similar in both developed and developing countries, mortality due to ARI is 10-50 times higher in developing countries. Viruses are common causes of ARI among such children, yet the disease burden of these infections in rural communities is unknown. METHODOLOGY/PRINCIPAL FINDINGS: A prospective longitudinal study was carried out in children enrolled from two rural Indian villages at birth and followed weekly for the development of ARI, classified as upper respiratory infection, acute lower respiratory infection (ALRI), or severe ALRI. Respiratory syncytial virus (RSV), influenza, parainfluenza viruses and adenoviruses in nasopharyngeal aspirates were detected by direct fluorescent antibody testing (DFA) and, in addition, centrifugation enhanced culture for RSV was done. 281 infants enrolled in 39 months and followed until 42 months. During 440 child years of follow-up there were 1307 ARIs, including 236 ALRIs and 19 severe ALRIs. Virus specific incidence rates per 1000 child years for RSV were total ARI 234, ALRI 39, and severe ALRI 9; for influenza A total ARI 141, ALRI 39; for INF B total ARI 37; for PIV1 total ARI 23, for PIV2 total ARI 28, ALRI 5; for parainfluenza virus 3 total ARI 229, ALRI 48, and severe ALRI 5 and for adenovirus total ARI 18, ALRI 5. Repeat infections with RSV were seen in 18 children. CONCLUSIONS/SIGNIFICANCE: RSV, influenza A and parainfluenza virus 3 were important causes of ARI among children in rural communities in India. These data will be useful for vaccine design, development and implementation purposes.


Assuntos
Infecções Respiratórias/epidemiologia , Infecções Respiratórias/virologia , Viroses/diagnóstico , Viroses/virologia , Vírus/isolamento & purificação , Doença Aguda , Adulto , Pré-Escolar , Estudos de Coortes , Feminino , Seguimentos , Humanos , Índia/epidemiologia , Lactente , Recém-Nascido , Estudos Longitudinais , Masculino , Nasofaringe/virologia , Reação em Cadeia da Polimerase , Estudos Prospectivos , População Rural , Vírus/genética , Adulto Jovem
9.
Am J Physiol Lung Cell Mol Physiol ; 293(2): L281-9, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17435077

RESUMO

Respiratory syncytial virus (RSV) is the most common cause of bronchiolitis in infants and children worldwide. We wished to determine whether intratracheal administration of beta-agonists improved alveolar fluid clearance (AFC) across the distal respiratory epithelium of RSV-infected mice. Following intranasal infection with RSV strain A2, AFC was measured in anesthetized, ventilated BALB/c mice by instillation of 5% BSA into the dependent lung. We found that direct activation of protein kinase A by forskolin or 8-bromo-cAMP increased AFC at day 2 after infection with RSV. In contrast, short- and long-acting beta-agonists had no effect at either day 2 or day 4. Insensitivity to beta-agonists was not a result of elevated plasma catecholamines or lung epithelial cell beta-adrenergic receptor degradation. Instead, RSV-infected mice had significantly higher levels of phosphorylated PKCzeta in the membrane fractions of their lung epithelial cells. In addition, insensitivity to beta-agonists was mediated in a paracrine fashion by KC (the murine homolog of CXCL8) and reversed by inhibition of either PKCzeta or G protein-coupled receptor kinase 2 (GRK2). These results indicate that insufficient response to beta-agonists in RSV may be caused, at least in part, by impaired beta-adrenergic receptor signaling, as a consequence of GRK2-mediated uncoupling of beta-adrenergic receptors from adenylyl cyclase.


Assuntos
Receptores Adrenérgicos beta 2/metabolismo , Mucosa Respiratória/metabolismo , Mucosa Respiratória/virologia , Infecções por Vírus Respiratório Sincicial/metabolismo , Vírus Sincicial Respiratório Humano/metabolismo , Agonistas de Receptores Adrenérgicos beta 2 , Agonistas Adrenérgicos beta/farmacologia , Animais , Catecolaminas/metabolismo , Quimiocina CXCL1 , Quimiocinas CXC/farmacologia , Colforsina/farmacologia , Água Extravascular Pulmonar/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/virologia , Organismos Livres de Patógenos Específicos , Terbutalina/farmacologia
10.
J Virol ; 80(23): 11651-7, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17005645

RESUMO

Respiratory syncytial virus (RSV) is the only infectious disease for which a monoclonal antibody (MAb) is used in humans. Palivizumab (PZ) is a humanized murine MAb to the F protein of RSV. PZ-resistant viruses appear after in vitro and in vivo growth of RSV in the presence of PZ. Fitness for replication could be a determinant of the likelihood of dissemination of resistant viruses. We assessed the fitness of two PZ-resistant viruses (F212 and MP4). F212 grew less well in cell culture than the parent A2 virus and was predicted to be less fit than A2. Equal amounts of F212 and A2 were mixed and passaged in cell culture. F212 disappeared from the viral population, indicating it was less fit than the A2 virus. The MP4 virus grew as well as A2 in culture and in cotton rats. A2/MP4 virus input ratios of 1:1, 10:1, 100:1, and 1,000:1 were compared in competitive replication. For all input ratios except 1,000:1, the MP4 virus became dominant, supplanting the A2 virus. The MP4 virus also dominated the A2 virus during growth in cotton rats. Thus, the mutant MP4 virus was more fit than A2 virus in both in vitro and in vivo competitive replication. Whether this fitness difference was due to the identified nucleotide substitutions in the F gene or to mutations elsewhere in the genome is unknown. Understanding the mechanisms by which mutant virus fitness increased or decreased could prove useful for consideration in attenuated vaccine design efforts.


Assuntos
Mutação , Vírus Sinciciais Respiratórios/fisiologia , Replicação Viral , Animais , Anticorpos Monoclonais/farmacologia , Linhagem Celular , RNA Viral/genética , RNA Viral/isolamento & purificação , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/imunologia , Sigmodontinae
11.
J Clin Microbiol ; 44(9): 3055-64, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16954227

RESUMO

Respiratory syncytial virus (RSV) is the most commonly identified viral agent of acute respiratory tract infection (ARI) of young children and causes repeat infections throughout life. Limited data are available on the molecular epidemiology of RSV from developing countries, including India. This study reports on the genetic variability in the glycoprotein G gene among RSV isolates from India. Reverse transcription-PCR for a region of the RSV G protein gene was done with nasopharyngeal aspirates (NPAs) collected in a prospective longitudinal study in two rural villages near Delhi and from children with ARI seen in an urban hospital. Nucleotide sequence comparisons among 48 samples demonstrated a higher prevalence of group A (77%) than group B (23%) RSV isolates. The level of genetic variability was higher among the group A viruses (up to 14%) than among the group B viruses (up to 2%). Phylogenetic analysis revealed that both the GA2 and GA5 group A RSV genotypes were prevalent during the 2002-2003 season and that genotype GA5 was predominant in the 2003-2004 season, whereas during the 2004-2005 season both genotype GA5 and genotype BA, a newly identified group B genotype, cocirculated in almost equal proportions. Comparison of the nonsynonymous mutation-to-synonymous mutation ratios (dN/dS) revealed greater evidence for selective pressure between the GA2 and GA5 genotypes (dN/dS, 1.78) than within the genotypes (dN/dS, 0.69). These are among the first molecular analyses of RSV strains from the second most populous country in the world and will be useful for comparisons to candidate vaccine strains.


Assuntos
Variação Genética , Infecções por Vírus Respiratório Sincicial/epidemiologia , Vírus Sincicial Respiratório Humano/classificação , Infecções Respiratórias/epidemiologia , Proteínas Virais de Fusão/genética , Doença Aguda , Sequência de Aminoácidos , Pré-Escolar , Hospitais Urbanos , Humanos , Índia/epidemiologia , Lactente , Recém-Nascido , Epidemiologia Molecular , Dados de Sequência Molecular , Nasofaringe/virologia , Filogenia , Prevalência , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/genética , Infecções Respiratórias/virologia , População Rural , Estações do Ano , Análise de Sequência de DNA
12.
Contemp Clin Trials ; 27(3): 254-9, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16603417

RESUMO

Delays in research on emerging infections could deprive the public of appropriate therapies. This report describes challenges encountered in implementing two multicenter protocols of West Nile virus (WNV) infections in the United States during 2003. Protocol development times, federal regulatory approvals, and local Institutional Review Boards (IRB) approvals were compiled. Twenty eight institutions participated in a natural history study and 27 in a therapeutic trial of WNV developed through the National Institute of Allergy and Infectious Disease Collaborative Antiviral Study Group (CASG). The CASG compiled protocol development times, federal regulatory approvals, and local IRB approvals. Additional information on the local IRB process was obtained by survey of the investigators. Because of the lengthy development and approval process, protocols were distributed after the start of the epidemic season, most sites were unable to enroll subjects at the peak of the season, and a number of sites lacked IRB approval at the end of the season.


Assuntos
Protocolos Clínicos/normas , Ensaios Clínicos como Assunto/normas , Febre do Nilo Ocidental/prevenção & controle , Ensaios Clínicos como Assunto/métodos , Progressão da Doença , Comitês de Ética em Pesquisa , Humanos , Incidência , Estudos Multicêntricos como Assunto , Estações do Ano , Fatores de Tempo , Estados Unidos/epidemiologia , Febre do Nilo Ocidental/epidemiologia
13.
J Med Virol ; 78(5): 659-65, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16555275

RESUMO

Respiratory syncytial virus (RSV) causes repeat infections throughout life. Antigenic variability in the RSV G protein may play a significant role in reinfections. A variable region of the RSV G gene was analyzed for 14 viruses from seven children who experienced initial and repeat infections. Eleven group A strains were in clades GA2 and GA5 and the three group B viruses were in the newly identified BA clade. In five children reinfections were caused by a heterologous group or genotype of RSV. Two children experienced infection and reinfection by viruses of the same clade, these virus pairs differed by only two to three amino acids in the region compared. This is the first report of RSV nucleotide sequence analysis from India and one of the few molecular characterizations of paired RSV from reinfections. Determining the molecular basis of reinfections may have important implications for RSV vaccine development.


Assuntos
Genes Virais , Variação Genética , Recidiva , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/genética , Proteínas Virais de Fusão/genética , Sequência de Aminoácidos , Pré-Escolar , Feminino , Humanos , Índia , Lactente , Estudos Longitudinais , Masculino , Dados de Sequência Molecular , Vírus Sinciciais Respiratórios/classificação , População Rural , Alinhamento de Sequência , Análise de Sequência , Especificidade da Espécie
14.
Am J Respir Crit Care Med ; 173(6): 639-43, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16387798

RESUMO

RATIONALE: Recently, respiratory syncytial virus (RSV) RNA has been identified by reverse transcriptase-polymerase chain reaction (RT-PCR) from a high percentage of patients with stable chronic obstructive pulmonary disease (COPD). These data raise the possibility of persistent low-grade infection in this population, which could have implications in COPD pathogenesis. OBJECTIVES: RSV persistence was investigated by testing respiratory secretions from subjects with COPD during illness and at regular intervals over 1 yr. METHODS: Nasal and sputum samples from subjects with COPD were tested by one-tube nested RT-PCR for RSV every 2 mo and during respiratory illnesses for 1 yr. Subjects positive for RSV were evaluated weekly until negative in two consecutive samples. Nasal secretions and serum were tested for RSV antibody. A rise of fourfold or greater was defined as evidence of RSV infection. RESULTS: A total of 112 patients were enrolled and the illnesses of 92 patients were evaluated. RSV was detected by RT-PCR in 6/92 (6.5%) illness nasal samples versus 0/685 routine nasal samples and in 5/69 (7.2%) illness sputum samples versus 3 /315 (0.9%) routine. Four additional RSV infections were identified by serum antibody responses. Of the RSV infections 86% were associated with serum or nasal antibody responses and 73% had symptoms of acute respiratory illness. CONCLUSIONS: Most RSV infections in patients with COPD are associated with symptomatic respiratory illnesses and measurable immune responses. Our data do not support the concept of RSV persistence in this population.


Assuntos
Anticorpos Antivirais/análise , Doença Pulmonar Obstrutiva Crônica/virologia , RNA Viral/análise , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/isolamento & purificação , Adulto , Idoso , Diagnóstico Diferencial , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Mucosa Nasal/virologia , Doença Pulmonar Obstrutiva Crônica/diagnóstico , Infecções por Vírus Respiratório Sincicial/diagnóstico , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/imunologia , Estudos Retrospectivos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Am J Respir Crit Care Med ; 173(6): 673-82, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16387801

RESUMO

RATIONALE: Previously, we demonstrated that intranasal infection of BALB/c mice with respiratory syncytial virus (RSV) resulted in an early 40% reduction in alveolar fluid clearance (AFC), an effect mediated via P2Y purinergic receptors. OBJECTIVES: To confirm that RSV-induced inhibition of AFC is mediated by uridine triphosphate (UTP), and to demonstrate that inhibition of de novo pyrimidine synthesis with leflunomide prevents increased UTP release after RSV infection, and thereby also prevents inhibition of AFC by RSV. METHODS: BALB/c mice were infected intranasally with RSV strain A2. AFC was measured in anesthetized, ventilated mice by instillation of 5% bovine serum albumin into the dependent lung. Some mice were pretreated with leflunomide or 6-mercaptopurine. MEASUREMENTS AND MAIN RESULTS: RSV-mediated inhibition of AFC is associated temporally with a 20-nM increase in UTP and ATP content of bronchoalveolar lavage fluid, hypoxemia, and altered nasal potential difference. RSV-mediated nucleotide release, AFC inhibition, and physiologic sequelae thereof can be prevented by pretreatment of mice with the de novo pyrimidine synthesis inhibitor leflunomide, which is not toxic to the mice, and which does not affect RSV replication in the lungs. In contrast, pretreatment of mice with 6-mercaptopurine, an inhibitor of de novo purine synthesis, has no beneficial effect on AFC or other indicators of disease progression. Finally, RSV-mediated inhibition of AFC is prevented by volume-regulated anion channel inhibitors. CONCLUSION: Pyrimidine synthesis or release pathways may provide novel therapeutic targets to counter the pathophysiologic sequelae of impaired AFC in RSV disease.


Assuntos
Antivirais/farmacologia , Líquido da Lavagem Broncoalveolar/virologia , Isoxazóis/farmacologia , Pneumonia Viral/tratamento farmacológico , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Vírus Sinciciais Respiratórios/patogenicidade , Animais , Modelos Animais de Doenças , Progressão da Doença , Feminino , Leflunomida , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Nucleotidases/metabolismo , Pneumonia Viral/metabolismo , Pneumonia Viral/virologia , Infecções por Vírus Respiratório Sincicial/metabolismo , Infecções por Vírus Respiratório Sincicial/virologia , Índice de Gravidade de Doença
16.
J Virol ; 79(7): 3962-8, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15767398

RESUMO

Palivizumab (PZ) is the only monoclonal antibody currently available for use in humans against an infectious disease. PZ is administered prophylactically for respiratory syncytial virus (RSV) infections. RSV selected in cell culture for growth in the presence of PZ develops F gene mutations and can be resistant to PZ prophylaxis in cotton rats. Here, we evaluated the potential for PZ-resistant RSV mutants to arise in vivo. Cotton rats were immunosuppressed with cyclophosphamide, administered PZ, and inoculated intranasally with RSV. Lungs were harvested 12 weeks after RSV infection, reverse transcription-PCR-amplified F gene fragments were cloned into plasmids, and the nucleotide sequences of the cloned cDNAs were determined. Three of the five animals had mixed populations of lung virus, and over 50% of the clones from the three animals revealed F gene mutations associated with resistance to PZ. A virus completely resistant to PZ neutralization was recovered from the lung homogenate of a rat that had received PZ. Thus, prolonged pulmonary replication of RSV in the presence of PZ was followed by the appearance of viruses resistant to PZ. The potential for the development of resistance is a consideration as the antibody is used prophylactically against RSV and as passively administered antibodies are under development for other infections, including emerging viruses and agents of biodefense.


Assuntos
Anticorpos Monoclonais/farmacologia , Antivirais/farmacologia , Farmacorresistência Viral , Pulmão/virologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Vírus Sinciciais Respiratórios/imunologia , Animais , Anticorpos Monoclonais Humanizados , Células Cultivadas , Análise Mutacional de DNA , DNA Complementar/química , DNA Complementar/isolamento & purificação , Humanos , Palivizumab , Mutação Puntual , RNA Viral/genética , RNA Viral/isolamento & purificação , Ratos , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/genética , Seleção Genética , Análise de Sequência de DNA , Sigmodontinae , Proteínas Virais/genética , Proteínas Virais/imunologia
17.
J Infect Dis ; 190(11): 1941-6, 2004 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15529258

RESUMO

BACKGROUND: Palivizumab (PZ) is the only monoclonal antibody in use against a human infectious disease. PZ is given as prophylaxis against infection with respiratory syncytial virus (RSV). An RSV escape mutant, MP4, has been shown to resist PZ prophylaxis in cotton rats. METHODS: To further define the potential of RSV to resist prophylaxis, additional PZ-resistant viruses were selected in cell culture and were tested for susceptibility to PZ in cotton rats. RESULTS: Mutant MS412 had an A-->C mutation at nucleotide position 827 in the F gene, resulting in an amino acid change from Lys to Gln at position 272. Mutant F212 had an A-->T mutation at position 816, leading to an amino acid change from Asn to Ile at position 268. In vitro, F212 had impaired growth kinetics. In cell culture, F212 was partially and MS412 was completely resistant to PZ neutralization. A single prophylactic dose of 15 mg/kg PZ protected cotton rats from infection with F212 but not with MS412. CONCLUSION: Both in vitro and in vivo, individual RSV PZ escape mutants varied in their susceptibility to PZ. Mutations associated with resistance to PZ did not always result in failure of PZ prophylaxis.


Assuntos
Anticorpos Monoclonais/farmacologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Substituição de Aminoácidos , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados , Farmacorresistência Viral/imunologia , Injeções Intramusculares , Mutação , Testes de Neutralização , Palivizumab , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/imunologia , Sigmodontinae , Proteínas Virais de Fusão/genética
18.
Virology ; 327(1): 121-33, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15327903

RESUMO

Sequences at the beginnings and ends of Human respiratory syncytial virus (HRSV) genes are necessary for efficient initiation and termination of transcription. The gene start sequences are well conserved and contain signals required for initiation, while the semi-conserved sequences at the gene ends direct transcriptional termination with varying efficiencies. The intergenic regions, which lie between the gene ends and the downstream gene start sequences, are not conserved in length or sequence, and certain positions have been reported to play a role in transcriptional regulation. We have previously shown that the gene end sequences in HRSV subgroup A clinical isolates are variable and that variations found at certain gene ends decreased transcriptional termination and downstream mRNA expression. Here, we have extended this work to examine variation in the intergenic regions between the genes of clinical isolates. We determined the sequences of the eight intergenic regions and the M2/L overlap from clinical isolates from the US and UK and found that all of these regions contained variations from the prototype A2 strain. The amount of variation observed was disparate among the different intergenic regions and did not correlate with length. The effects of selected variant sequences on transcription were examined in the context of subgenomic replicons. While some changes in the intergenic regions had minor effects, certain sequence variations significantly altered transcription termination or initiation. A single nucleotide deletion in the M/SH intergenic region decreased initiation at the SH gene start seven-fold, while changes in the F/M2 intergenic region were found that in some cases increased and in others decreased termination at the F gene end. The P/M intergenic region was the most variable, but none of the changes examined affected either termination at the P gene end or initiation of the downstream M gene start. These results show that in HRSV clinical isolates the intergenic region sequences are variable and that changes in these regions have the potential to affect transcriptional control at the gene junctions.


Assuntos
DNA Intergênico/genética , Regulação Viral da Expressão Gênica , Variação Genética , Vírus Sincicial Respiratório Humano/genética , Transcrição Gênica , Sequência de Bases , Humanos , Dados de Sequência Molecular , RNA Viral/genética , Vírus Sincicial Respiratório Humano/classificação , Análise de Sequência de DNA , Deleção de Sequência , Proteínas Virais/genética , Proteínas Virais/metabolismo
19.
Virology ; 318(2): 608-12, 2004 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-14972528

RESUMO

Palivizumab (PZ) is the only monoclonal antibody in human use against an infectious disease. PZ is a humanized monoclonal antibody that recognizes the fusion protein of respiratory syncytial virus (RSV). PZ prophylaxis reduces the likelihood of hospitalization for young children at risk for severe RSV infections. The quasispecies nature of RNA viruses allows rapid emergence of viruses with a selective advantage. A PZ resistant virus was selected by passage of RSV in the presence of PZ in cell culture. The cell culture-derived virus was completely resistant to PZ prophylaxis in cotton rats. The increasing use of PZ, and in particular, the use of PZ in immunosuppressed patients, provide opportunities for resistant viruses to emerge. Whether such viruses will appear and be of clinical significance for humans is unknown. Preclinical studies in cotton rats predicted the efficacy of PZ in humans; these results suggest that if PZ resistant viruses arise in humans, PZ prophylaxis may be ineffective.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antivirais/uso terapêutico , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sinciciais Respiratórios , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Antivirais/farmacologia , Linhagem Celular , Modelos Animais de Doenças , Humanos , Mutação , Palivizumab , Ratos , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Vírus Sinciciais Respiratórios/genética , Sigmodontinae
20.
Am J Physiol Lung Cell Mol Physiol ; 286(1): L112-20, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12948936

RESUMO

Respiratory syncytial virus (RSV) is the most common cause of lower respiratory tract disease in infants and children worldwide. Intranasal infection of BALB/c mice with RSV strain A2, but not ultraviolet-inactivated RSV, for 2 or 4 days reduced basal alveolar fluid clearance (AFC), a seminal function of bronchoalveolar epithelium, and caused loss of AFC sensitivity to amiloride inhibition. Reduced AFC was temporally associated with increased lung water content but was not a consequence of increased epithelial permeability or cell death. Reduced AFC was also not due to decreased transcription of epithelial Na+ channel subunit genes in lung tissue. RSV-mediated inhibition of AFC 2 days after infection was rapidly prevented by addition to the instillate of P2Y receptor antagonists (suramin and XAMR-0721) or enzymes that degrade UTP, but not those that degrade ATP. After UTP degradation, AFC returned to control levels but was no longer sensitive to amiloride. UTP at nanomolar concentrations recapitulated the AFC inhibitory effect of RSV in normal mice and mice infected with RSV for 6 days, indicating that normalization of AFC at this time point is a consequence of cessation of UTP release, rather than P2Y receptor desensitization. We conclude that RSV infection of the bronchoalveolar epithelium results in reduced AFC as a consequence of autocrine feedback inhibition mediated by UTP. These studies are the first to demonstrate AFC inhibition by an important pulmonary viral pathogen. Reduced AFC may result in formation of an increased volume of fluid mucus, airway congestion, and rhinorrhea, all features of severe RSV disease.


Assuntos
Água Extravascular Pulmonar/metabolismo , Alvéolos Pulmonares/metabolismo , Infecções por Vírus Respiratório Sincicial/metabolismo , Uridina Trifosfato/metabolismo , Animais , Peso Corporal , Canais Epiteliais de Sódio , Retroalimentação Fisiológica , Feminino , Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Alvéolos Pulmonares/virologia , Antagonistas do Receptor Purinérgico P2 , Receptores Purinérgicos P2/metabolismo , Sódio/metabolismo , Canais de Sódio/genética , Canais de Sódio/metabolismo , Uridina Trifosfato/farmacologia , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...