Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Viruses ; 14(5)2022 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-35632652

RESUMO

RNA viruses like SARS-CoV-2, influenza virus, and respiratory syncytial virus (RSV) are dependent on host genes for replication. We investigated if probenecid, an FDA-approved and safe urate-lowering drug that inhibits organic anion transporters (OATs) has prophylactic or therapeutic efficacy to inhibit RSV replication in three epithelial cell lines used in RSV studies, i.e., Vero E6 cells, HEp-2 cells, and in primary normal human bronchoepithelial (NHBE) cells, and in BALB/c mice. The studies showed that nanomolar concentrations of all probenecid regimens prevent RSV strain A and B replication in vitro and RSV strain A in vivo, representing a potential prophylactic and chemotherapeutic for RSV.


Assuntos
COVID-19 , Infecções por Vírus Respiratório Sincicial , Vírus Sincicial Respiratório Humano , Animais , Camundongos , Probenecid/farmacologia , Probenecid/uso terapêutico , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Infecções por Vírus Respiratório Sincicial/metabolismo , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sincicial Respiratório Humano/genética , SARS-CoV-2 , Replicação Viral
2.
J Virol ; 95(15): e0069221, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-33980596

RESUMO

Swine influenza virus (SIV) can cause respiratory illness in swine. Swine contribute to influenza virus reassortment, as avian, human, and/or swine influenza viruses can infect swine and reassort, and new viruses can emerge. Thus, it is important to determine the host antiviral responses that affect SIV replication. In this study, we examined the innate antiviral cytokine response to SIV by swine respiratory epithelial cells, focusing on the expression of interferon (IFN) and interferon-stimulated genes (ISGs). Both primary and transformed swine nasal and tracheal respiratory epithelial cells were examined following infection with field isolates. The results show that IFN and ISG expression is maximal at 12 h postinfection (hpi) and is dependent on cell type and virus genotype. IMPORTANCE Swine are considered intermediate hosts that have facilitated influenza virus reassortment events that have given rise pandemics or genetically related viruses have become established in swine. In this study, we examine the innate antiviral response to swine influenza virus in primary and immortalized swine nasal and tracheal epithelial cells, and show virus strain- and host cell type-dependent differential expression of key interferons and interferon-stimulated genes.


Assuntos
Citocinas/metabolismo , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H1N2/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Mucosa Respiratória/imunologia , Animais , Linhagem Celular , Citocinas/imunologia , Cães , Células Epiteliais/virologia , Interações Hospedeiro-Patógeno/imunologia , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H1N2/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H3N2/crescimento & desenvolvimento , Interferons/imunologia , Células Madin Darby de Rim Canino , Infecções por Orthomyxoviridae/imunologia , Mucosa Respiratória/citologia , Suínos , Replicação Viral/fisiologia
3.
J Virol Methods ; 288: 114015, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33271254

RESUMO

Detection of swine influenza virus (SIV) in commercial swine herds is important for understanding the infection status of the herd and for controlling disease. Current molecular diagnostics require that specimens be submitted to a laboratory which provides results to the growers after some time which is generally too late to intercede in disease control. Moreover, current diagnostic assays are time-consuming, typically costly, and require skilled technical expertise. We have instituted a reverse transcription loop-mediated isothermal amplification (RT-LAMP) diagnostic assay based on conserved regions of the SIV matrix (M) gene and H1N1 hemagglutinin (HA) sequences. The RT-LAMP assay was optimized to use both colorimetric and fluorescent endpoints and was validated. The M and HA RT-LAMP assays have a limit-of-detection (LOD) sensitive to 11 and 8-log-fold dilutions of viral RNA, respectively, and are capable of discriminating between H1 and H3 strains of SIV. Additionally, the RT-LAMP assay was optimized for direct amplification of SIV from field samples without the need for viral RNA isolation. The direct RT-LAMP detected >86 % of qRT-PCR validated SIV samples, and >66 % of negative samples when spiked with viral RNA or SIV. The diagnostic RT-LAMP assay is a rapid, sensitive, specific, and cost-effective method for the detection of SIV in herds substantially aiding diagnosis and surveillance.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Vírus da Influenza A , Animais , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A/genética , Técnicas de Diagnóstico Molecular , Técnicas de Amplificação de Ácido Nucleico , Transcrição Reversa , Sensibilidade e Especificidade , Suínos
4.
Vet Microbiol ; 251: 108914, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33181438

RESUMO

Tracking the genetic diversity and spread of swine influenza viruses (SIVs) in commercial swine farms is central for control and to reduce the potential emergence of SIV reassortants. We analyzed the diversity of SIVs in nasal washes or oral fluids from commercial swine farms in North Carolina using influenza M qRT-PCR and hemagglutinin (HA) and neuraminidase (NA) subtyping. We found a predominance of H1 HAs and N2 NAs in the samples examined. The majority of the H1 HAs could be further classified into gamma and delta subclusters. We also identified HAs of the H1 alpha cluster, and those of human novel pandemic origin. Glycan binding profiles from a representative subset of these viruses revealed broad α2,6 sialylated glycan recognition, though some strains exhibited the ability to bind to α2,3 sialic acid. These data show that SIV surveillance can aid our understanding of viral transmission dynamics and help uncover the diversity at the human-swine interface.


Assuntos
Fazendas/estatística & dados numéricos , Infecções por Orthomyxoviridae/epidemiologia , Infecções por Orthomyxoviridae/veterinária , Orthomyxoviridae/genética , Doenças dos Suínos/epidemiologia , Animais , Variação Genética , Hemaglutininas Virais/genética , Humanos , Meio-Oeste dos Estados Unidos/epidemiologia , Neuraminidase/genética , Orthomyxoviridae/classificação , Infecções por Orthomyxoviridae/transmissão , Filogenia , RNA Viral/genética , Vírus Reordenados/genética , Sudeste dos Estados Unidos/epidemiologia , Suínos , Doenças dos Suínos/virologia , Proteínas Virais/genética
6.
Vet Microbiol ; 211: 174-179, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29102115

RESUMO

Swine influenza A virus (IAV) can cause widespread respiratory disease with high morbidity, low mortality, and have a substantial economic impact to the swine industry. Swine infection may contribute to pandemic IAV given their susceptibility to both avian and human IAVs. Currently, three IAV subtypes (H1N1, H3N2 and H1N2) circulate in swine in North America frequently combining gene segments from avian or human viruses. This study investigated the prevalence of IAV in commercial swine herds. A total of 1878 oral fluid samples were collected from pigs of all ages from 201 commercial farms located in North Carolina and South Carolina. Sixty-eight oral fluid samples from 35 farms were positive by MP gene PCR with an overall IAV-positivity of 3.6%. On the herd level, the percentage of IAV positivity was 17.4%. Fifty-six viruses were subtyped, while 12 were partly subtyped or not subtyped at all. Using de novo assembly, complete sequences were obtained for 59 HA genes. The majority of IAVs subtyped had an H1 HA demonstrating a considerable prevalence over H3 viruses. Furthermore, only six out of eleven HA types were detected which has implications for the selection of vaccines used by swine producers in the region.


Assuntos
Vírus da Influenza A Subtipo H1N1/isolamento & purificação , Vírus da Influenza A Subtipo H1N2/isolamento & purificação , Vírus da Influenza A Subtipo H3N2/isolamento & purificação , Infecções por Orthomyxoviridae/veterinária , Animais , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N2/genética , Vírus da Influenza A Subtipo H3N2/genética , Epidemiologia Molecular , North Carolina/epidemiologia , Infecções por Orthomyxoviridae/epidemiologia , Infecções por Orthomyxoviridae/virologia , Filogenia , South Carolina/epidemiologia , Suínos
7.
ACS Appl Mater Interfaces ; 8(17): 10761-6, 2016 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-27064729

RESUMO

Carbon dots, generally defined as small carbon nanoparticles with various surface passivation schemes, have emerged as a new class of quantum-dot-like nanomaterials, with their optical properties and photocatalytic functions resembling those typically found in conventional nanoscale semiconductors. In this work, carbon dots were evaluated for their photoinduced bactericidal functions, with the results suggesting that the dots were highly effective in bacteria-killing with visible-light illumination. In fact, the inhibition effect could be observed even simply under ambient room lighting conditions. Mechanistic implications of the results are discussed and so are opportunities in the further development of carbon dots into a new class of effective visible/natural light-responsible bactericidal agents for a variety of bacteria control applications.


Assuntos
Pontos Quânticos , Carbono , Luz , Nanoestruturas , Semicondutores
8.
J Virol ; 90(4): 1694-704, 2016 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-26581994

RESUMO

UNLABELLED: Vaccine manufacturing costs prevent a significant portion of the world's population from accessing protection from vaccine-preventable diseases. To enhance vaccine production at reduced costs, a genome-wide RNA interference (RNAi) screen was performed to identify gene knockdown events that enhanced poliovirus replication. Primary screen hits were validated in a Vero vaccine manufacturing cell line using attenuated and wild-type poliovirus strains. Multiple single and dual gene silencing events increased poliovirus titers >20-fold and >50-fold, respectively. Host gene knockdown events did not affect virus antigenicity, and clustered regularly interspaced short palindromic repeat (CRISPR)-Cas9-mediated knockout of the top candidates dramatically improved viral vaccine strain production. Interestingly, silencing of several genes that enhanced poliovirus replication also enhanced replication of enterovirus 71, a clinically relevant virus to which vaccines are being targeted. The discovery that host gene modulation can markedly increase virus vaccine production dramatically alters mammalian cell-based vaccine manufacturing possibilities and should facilitate polio eradication using the inactivated poliovirus vaccine. IMPORTANCE: Using a genome-wide RNAi screen, a collection of host virus resistance genes was identified that, upon silencing, increased poliovirus and enterovirus 71 production by from 10-fold to >50-fold in a Vero vaccine manufacturing cell line. This report provides novel insights into enterovirus-host interactions and describes an approach to developing the next generation of vaccine manufacturing through engineered vaccine cell lines. The results show that specific gene silencing and knockout events can enhance viral titers of both attenuated (Sabin strain) and wild-type polioviruses, a finding that should greatly facilitate global implementation of inactivated polio vaccine as well as further reduce costs for live-attenuated oral polio vaccines. This work describes a platform-enabling technology applicable to most vaccine-preventable diseases.


Assuntos
Poliomielite/prevenção & controle , Vacinas contra Poliovirus/isolamento & purificação , Poliovirus/isolamento & purificação , Poliovirus/fisiologia , Tecnologia Farmacêutica/métodos , Replicação Viral , Animais , Vacinas Atenuadas/isolamento & purificação , Células Vero , Cultura de Vírus/métodos
9.
J Virol ; 87(19): 10792-804, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23903841

RESUMO

Influenza A virus and respiratory syncytial virus (RSV) cause substantial morbidity and mortality afflicting the ends of the age spectrum during the autumn through winter months in the United States. The benefit of vaccination against RSV and influenza using a subunit vaccine to enhance immunity and neutralizing antibody was investigated. Influenza virus hemagglutinin (HA) and RSV fusion (F) protein were tested as vaccine components alone and in combination to explore the adjuvant properties of RSV F protein on HA immunity. Mice vaccinated with HA and F exhibited robust immunity that, when challenged, had reduced viral burden for both influenza and RSV. These studies show an enhancing and cross-protective benefit of F protein for anti-HA immunity.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Vírus da Influenza A/patogenicidade , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/uso terapêutico , Vírus Sinciciais Respiratórios/patogenicidade , Proteínas Virais de Fusão/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Western Blotting , Células Cultivadas , Chlorocebus aethiops , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Humanos , Vírus da Influenza A/imunologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Subunidades Proteicas , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/imunologia , Receptor 4 Toll-Like/fisiologia , Células Vero , Proteínas Virais de Fusão/genética
10.
J Gen Virol ; 93(Pt 11): 2346-2356, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22894925

RESUMO

Respiratory syncytial virus (RSV) causes substantial morbidity and life-threatening lower respiratory tract disease in infants, young children and the elderly. Understanding the host response to RSV infection is critical for developing disease-intervention approaches. The role of microRNAs (miRNAs) in post-transcriptional regulation of host genes responding to RSV infection is not well understood. In this study, it was shown that RSV infection of a human alveolar epithelial cell line (A549) induced five miRNAs (let-7f, miR-24, miR-337-3p, miR-26b and miR-520a-5p) and repressed two miRNAs (miR-198 and miR-595), and showed that RSV G protein triggered let-7f expression. Luciferase-untranslated region reporters and miRNA mimics and inhibitors validated the predicted targets, which included cell-cycle genes (CCND1, DYRK2 and ELF4), a chemokine gene (CCL7) and the suppressor of cytokine signalling 3 gene (SOCS3). Modulating let-7 family miRNA levels with miRNA mimics and inhibitors affected RSV replication, indicating that RSV modulates host miRNA expression to affect the outcome of the antiviral host response, and this was mediated in part through RSV G protein expression.


Assuntos
Regulação Viral da Expressão Gênica/fisiologia , MicroRNAs/metabolismo , Processamento Pós-Transcricional do RNA/fisiologia , Vírus Sinciciais Respiratórios/fisiologia , Replicação Viral/fisiologia , Animais , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Humanos , MicroRNAs/genética , Alvéolos Pulmonares/citologia , Células Vero
11.
Viral Immunol ; 25(3): 193-203, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22551066

RESUMO

Respiratory syncytial virus (RSV) is a primary cause of severe lower respiratory tract disease in infants, young children, and the elderly worldwide, and despite decades of effort, there remains no safe and effective vaccine. RSV modifies the host immune response during infection by CX3C chemokine mimicry adversely affecting pulmonary leukocyte chemotaxis and CX3CR1+ RSV-specific T-cell responses. In this study we investigated whether immunization of mice with RSV G protein polypeptides from strain A2 could induce antibodies that block G protein-CX3CR1 interactions of both RSV A and B strains. The results show that mice immunized with RSV A2 G polypeptides generate antibodies that block binding of RSV A2 and B1 native G proteins to CX3CR1, and that these antibodies effectively cross-neutralize both A and B strains of RSV. These findings suggest that vaccines that induce RSV G protein-CX3CR1 blocking antibodies may provide a disease intervention strategy in the efforts to develop safe and efficacious RSV vaccines.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Quimiocinas CX3C/metabolismo , Ligação Proteica/efeitos dos fármacos , Receptores de Quimiocinas/metabolismo , Vírus Sincicial Respiratório Humano/imunologia , Proteínas Virais de Fusão/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Receptor 1 de Quimiocina CX3C , Linhagem Celular , Reações Cruzadas , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/classificação , Vacinação , Proteínas Virais de Fusão/administração & dosagem
12.
PLoS One ; 6(6): e21183, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21731666

RESUMO

Avian influenza viruses (AIV) are an important emerging threat to public health. It is thought that sialic acid (sia) receptors are barriers in cross-species transmission where the binding preferences of AIV and human influenza viruses are sias α2,3 versus α2,6, respectively. In this study, we show that a normal fully differentiated, primary human bronchial epithelial cell model is readily infected by low pathogenic H5N1, H5N2 and H5N3 AIV, which primarily bind to sia α2,3 moieties, and replicate in these cells independent of specific sias on the cell surface. NHBE cells treated with neuraminidase prior to infection are infected by AIV despite removal of sia α2,3 moieties. Following AIV infection, higher levels of IP-10 and RANTES are secreted compared to human influenza virus infection, indicating differential chemokine expression patterns, a feature that may contribute to differences in disease pathogenesis between avian and human influenza virus infections in humans.


Assuntos
Aves/virologia , Brônquios/patologia , Células Epiteliais/virologia , Vírus da Influenza A/fisiologia , Influenza Aviária/virologia , Influenza Humana/virologia , Ácido N-Acetilneuramínico/metabolismo , Adolescente , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Quimiocinas/metabolismo , Cílios/efeitos dos fármacos , Cílios/metabolismo , Cílios/patologia , Cães , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Células Caliciformes/efeitos dos fármacos , Células Caliciformes/metabolismo , Células Caliciformes/patologia , Humanos , Vírus da Influenza A/efeitos dos fármacos , Influenza Aviária/patologia , Influenza Humana/patologia , Masculino , Neuraminidase/farmacologia , Receptores de Superfície Celular/metabolismo , Replicação Viral/efeitos dos fármacos , Replicação Viral/fisiologia , Eliminação de Partículas Virais/efeitos dos fármacos , Eliminação de Partículas Virais/fisiologia
13.
J Virol ; 84(2): 1148-57, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19864390

RESUMO

Respiratory syncytial virus (RSV) infection causes substantial morbidity and some deaths in the young and elderly worldwide. There is no safe and effective vaccine available, although it is possible to reduce the hospitalization rate for high-risk children by anti-RSV antibody prophylaxis. RSV has been shown to modify the immune response to infection, a feature linked in part to RSV G protein CX3C chemokine mimicry. This study determined if vaccination with G protein polypeptides or peptides spanning the central conserved region of the G protein could induce antibodies that blocked G protein CX3C-CX3CR1 interaction and disease pathogenesis mediated by RSV infection. The results show that mice vaccinated with G protein peptides or polypeptides containing the CX3C motif generate antibodies that inhibit G protein CX3C-CX3CR1 binding and chemotaxis, reduce lung virus titers, and prevent body weight loss and pulmonary inflammation. The results suggest that RSV vaccines that induce antibodies that block G protein CX3C-CX3CR1 interaction may offer a new, safe, and efficacious RSV vaccine strategy.


Assuntos
Anticorpos Antivirais/sangue , Quimiocinas CX3C/metabolismo , Receptores de Quimiocinas/metabolismo , Infecções por Vírus Respiratório Sincicial , Vacinas contra Vírus Sincicial Respiratório , Proteínas Virais de Fusão/imunologia , Animais , Anticorpos Antivirais/imunologia , Receptor 1 de Quimiocina CX3C , Linhagem Celular , Quimiocinas CX3C/imunologia , Quimiotaxia de Leucócito/imunologia , Feminino , Humanos , Inflamação/imunologia , Inflamação/prevenção & controle , Pulmão/imunologia , Pulmão/fisiopatologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos/síntese química , Peptídeos/química , Peptídeos/imunologia , Receptores de Quimiocinas/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/fisiopatologia , Infecções por Vírus Respiratório Sincicial/virologia , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Vírus Sincicial Respiratório Humano/patogenicidade , Vírus Sincicial Respiratório Humano/fisiologia , Vacinação , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Replicação Viral
14.
Viral Immunol ; 22(3): 147-61, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19435411

RESUMO

Respiratory syncytial virus (RSV) is a common cause of repeat infections throughout life and potentially severe lower respiratory tract illness in infants, young children, and the elderly. RSV proteins have been shown to contribute to immune evasion by several means, including modification of cytokine and chemokine responses whose expression is negatively regulated by suppressor of cytokine signaling (SOCS) proteins. In this study, we examine the role of SOCS1 and SOCS3 regulation of the type I interferon (IFN) response in normal fully-differentiated human bronchial epithelial cells infected with RSV or with an RSV mutant virus lacking the G gene. The results show that RSV G protein modulates SOCS expression to inhibit type I IFN and interferon-stimulated gene (ISG)-15 expression very early as well as late in infection, and that SOCS induction is linked to toll-like receptor (TLR) signaling by RSV F protein, as indicated by interferon-regulatory factor (IRF)-3 activation and nuclear translocation. These findings indicate that RSV surface proteins signal through the TLR pathway, suggesting that this may be an important mechanism to reduce type I IFN expression to aid virus replication.


Assuntos
Interferon-alfa/biossíntese , Interferon beta/biossíntese , Infecções por Vírus Respiratório Sincicial/metabolismo , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/fisiologia , Proteínas Supressoras da Sinalização de Citocina/biossíntese , Receptores Toll-Like/metabolismo , Linhagem Celular , Citocinas/biossíntese , Citocinas/imunologia , Regulação da Expressão Gênica/imunologia , Humanos , Fator Regulador 3 de Interferon/biossíntese , Fator Regulador 3 de Interferon/imunologia , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferon beta/genética , Interferon beta/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Proteína 1 Supressora da Sinalização de Citocina , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/imunologia , Receptores Toll-Like/imunologia , Ativação Transcricional , Ubiquitinas/biossíntese , Ubiquitinas/imunologia , Proteínas Virais de Fusão/fisiologia , Replicação Viral
15.
J Immunol ; 176(3): 1600-8, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16424189

RESUMO

Interactions between fractalkine (CX3CL1) and its receptor, CX3CR1, mediate leukocyte adhesion, activation, and trafficking. The respiratory syncytial virus (RSV) G protein has a CX3C chemokine motif that can bind CX3CR1 and modify CXCL1-mediated responses. In this study, we show that expression of the RSV G protein or the G protein CX3C motif during infection is associated with reduced CX3CR1+ T cell trafficking to the lung, reduced frequencies of RSV-specific, MHC class I-restricted IFN-gamma-expressing cells, and lower numbers of IL-4- and CX3CL1-expressing cells. In addition, we show that CX3CR1+ cells constitute a major component of the cytotoxic response to RSV infection. These results suggest that G protein and the G protein CX3C motif reduce the antiviral T cell response to RSV infection.


Assuntos
Motivos de Aminoácidos , Quimiocinas CX3C/genética , Receptores de Quimiocinas/biossíntese , Vírus Sinciciais Respiratórios/imunologia , Linfócitos T/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Receptor 1 de Quimiocina CX3C , Inibição de Migração Celular , Movimento Celular/imunologia , Células Cultivadas , Quimiocinas CX3C/biossíntese , Feminino , Interferon gama/biossíntese , Interferon gama/genética , Interleucina-4/metabolismo , Pulmão/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Receptores de Quimiocinas/antagonistas & inibidores , Vírus Sinciciais Respiratórios/genética , Linfócitos T/metabolismo , Linfócitos T/patologia , Linfócitos T/virologia , Proteínas do Envelope Viral/biossíntese , Proteínas do Envelope Viral/genética
16.
J Virol Methods ; 128(1-2): 21-8, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15885812

RESUMO

Monoclonal antibodies (Mabs) against the Urbani strain of the SARS-associated coronavirus (SARS-CoV) were developed and characterized for reactivity to SARS-CoV and SARS-CoV S, N, M, and E proteins using enzyme-linked immunoabsorbent (ELISA), radioimmunoprecipitation, immunofluorescence, Western Blot and microneutralization assays. Twenty-six mAbs were reactive to SARS-CoV by ELISA, and nine were chosen for detailed characterization. Five mAbs reacted against the S protein, two against the M protein, and one each against the N and E proteins. Two of five S protein mAbs neutralized SARS-CoV infection of Vero E6 cells and reacted to an epitope within amino acids 490-510 in the S protein. While two of the three non-neutralizing antibodies recognized at second epitope within amino acids 270-350. The mAbs characterized should prove useful for developing SARS-CoV diagnostic assays and for studying the biology of infection and pathogenesis of disease.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Especificidade de Anticorpos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Proteínas Estruturais Virais/imunologia , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Antivirais/biossíntese , Linhagem Celular , Chlorocebus aethiops , Proteínas M de Coronavírus , Proteínas do Nucleocapsídeo de Coronavírus , Mapeamento de Epitopos , Humanos , Glicoproteínas de Membrana/imunologia , Testes de Neutralização , Proteínas do Nucleocapsídeo/imunologia , Síndrome Respiratória Aguda Grave/virologia , Glicoproteína da Espícula de Coronavírus , Células Vero , Proteínas do Envelope Viral/imunologia , Proteínas da Matriz Viral/imunologia , Proteínas Viroporinas
17.
Virus Res ; 105(1): 67-73, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15325082

RESUMO

Respiratory tract infections are a leading cause of morbidity and mortality worldwide. Human metapneumovirus (HMPV) is a recently discovered respiratory pathogen of the Paramyxovirus family in the Metapneumovirus genus. HMPV was first isolated from young children in The Netherlands with respiratory illness similar to human respiratory syncytial virus (RSV) infection. Epidemiological data indicates that HMPV co-circulates with RSV in the community. Few immunological tools are available to study the virological features of HMPV infection, thus current studies rely on reverse-transcription (RT) polymerase chain reaction (PCR) for detection. In this study, we examine serological cross-reactivity of RSV, HMPV and other Metapneumovirus members, i.e. avian metapneumovirus (AMPV), and show that polyclonal and monoclonal antibodies reactive to a conserved region in AMPV nucleoprotein (N) cross-react with HMPV N protein, but not with RSV N protein by ELISA, Western blot and immunohistochemical assays. In addition, we show that HMPV infection in the lungs of BALB/c mice can be detected using anti-N protein antibody. These reagents provide new tools and methods for investigating HMPV infection, for differentiating HMPV from RSV infection, and may be useful for characterizing potential links between HMPV with other respiratory complications.


Assuntos
Anticorpos Antivirais/imunologia , Metapneumovirus/imunologia , Metapneumovirus/isolamento & purificação , Proteínas do Nucleocapsídeo/imunologia , Infecções por Paramyxoviridae/diagnóstico , Infecções Respiratórias/diagnóstico , Animais , Anticorpos Monoclonais/imunologia , Antígenos Virais/análise , Antígenos Virais/imunologia , Western Blotting , Chlorocebus aethiops , Reações Cruzadas , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas do Nucleocapsídeo/análise , RNA Viral/isolamento & purificação , Vírus Sincicial Respiratório Humano/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Vero
18.
J Virol ; 77(18): 9831-44, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12941892

RESUMO

Vaccination with formalin-inactivated respiratory syncytial virus (FI-RSV) vaccine or RSV G glycoprotein results in enhanced pulmonary disease after live RSV infection. Enhanced pulmonary disease is characterized by pulmonary eosinophilia and is associated with a substantial inflammatory response. We show that the absence of the G glycoprotein or G glycoprotein CX3C motif during FI-RSV vaccination or RSV challenge of FI-RSV-vaccinated mice, or treatment with anti-substance P or anti-CX3CR1 antibodies, reduces or eliminates enhanced pulmonary disease, modifies T-cell receptor Vbeta usage, and alters CC and CXC chemokine expression. These data suggest that the G glycoprotein, and in particular the G glycoprotein CX3C motif, is key in the enhanced inflammatory response to FI-RSV vaccination, possibly through the induction of substance P.


Assuntos
Quimiocinas CX3C/metabolismo , Proteínas de Membrana , Eosinofilia Pulmonar/etiologia , Receptores de Quimiocinas/metabolismo , Vírus Sinciciais Respiratórios/imunologia , Substância P/biossíntese , Proteínas Virais/fisiologia , Vacinas Virais/toxicidade , Motivos de Aminoácidos , Animais , Linfócitos T CD4-Positivos/imunologia , Receptor 1 de Quimiocina CX3C , Movimento Celular , Quimiocinas/genética , Feminino , Formaldeído , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/análise , Receptores de Antígenos de Linfócitos T alfa-beta/fisiologia , Vacinação/efeitos adversos , Vacinas de Produtos Inativados/toxicidade , Proteínas Virais/química
19.
J Virol ; 77(11): 6580-4, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12743318

RESUMO

Respiratory syncytial virus (RSV) infection in the neonate can alter respiratory rates, i.e., lead to episodes of apnea. We show that RSV G glycoprotein reduces respiratory rates associated with the induction of substance P (SP) and G glycoprotein-CX3CR1 interaction, an effect that is inhibited by treatment with anti-G glycoprotein, anti-SP, or anti-CX3CR1 monoclonal antibodies. These data suggest new approaches for treating some aspects of RSV disease.


Assuntos
Receptores de Citocinas/metabolismo , Receptores de HIV/metabolismo , Respiração/efeitos dos fármacos , Vírus Sincicial Respiratório Humano/patogenicidade , Substância P/metabolismo , Proteínas Virais/fisiologia , Animais , Apneia/etiologia , Receptor 1 de Quimiocina CX3C , Quimiocinas CX3C/química , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Virais/química , Proteínas Virais/farmacologia
20.
Clin Diagn Lab Immunol ; 9(3): 633-8, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11986272

RESUMO

The role of strain differences in respiratory syncytial virus (RSV) disease has not been clearly defined. To investigate the possibility that strain differences contribute to susceptibility to repeat infections, we developed assays to detect antibodies to the two variable regions of the RSV G protein by cloning and expressing the internal variable region at amino acids (aa) 60 to 172 (g1) and the carboxy-terminal variable region at aa 193 to the carboxy terminus (g2) from different genotypes of RSV. The purified proteins were covalently linked to beads with different proportions of red and orange fluorescent dyes and reacted against serum specimens. Antibody reacting against the differently colored beads, and thus against different G polypeptides, was detected by use of flow cytometry and the Luminex system. This assay system detected group- and, to some extent, genotype-specific responses to RSV infection and can be used to investigate the role of strain differences in RSV disease.


Assuntos
Anticorpos Antivirais/sangue , Antígenos Virais/imunologia , Peptídeos/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/imunologia , Proteínas Virais/imunologia , Animais , Anticorpos Antivirais/imunologia , Pré-Escolar , Humanos , Lactente , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Vírus Respiratório Sincicial/sangue , Infecções por Vírus Respiratório Sincicial/diagnóstico , Infecções por Vírus Respiratório Sincicial/imunologia , Vacinas Atenuadas , Vacinas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...