Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Respir Cell Mol Biol ; 61(4): 450-458, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30916989

RESUMO

CDHR3 (cadherin-related family member 3) is a transmembrane protein that is highly expressed in airway epithelia and the only known receptor for rhinovirus C (RV-C). A CDHR3 SNP (rs6967330) with G to A base change has been linked to severe exacerbations of asthma and increased susceptibility to RV-C infections in young children. The goals of this study were to determine the subcellular localization of CDHR3 and to test the hypothesis that CDHR3 asthma-risk genotype affects epithelial cell function and susceptibility to RV-C infections of the airway epithelia. We used immunofluorescence imaging, Western blot analysis, and transmission electron microscopy to show CDHR3 subcellular localization in apical cells, including expression in the cilia of airway epithelia. Polymorphisms in CDHR3 rs6967330 locus (G→A) that were previously associated with childhood asthma were related to differences in CDHR3 expression and epithelial cell function. The rs6967330 A allele was associated with higher overall protein expression and RV-C binding and replication compared with the rs6967330 G allele. Furthermore, the rs6967330 A allele was associated with earlier ciliogenesis and higher FOXJ1 expression. Finally, CDHR3 genotype had no significant effects on membrane integrity or ciliary beat function. These findings provide information on the subcellular localization and possible functions of CDHR3 in the airways and link CDHR3 asthma-risk genotype to increased RV-C binding and replication.


Assuntos
Caderinas/genética , Células Epiteliais/virologia , Proteínas de Membrana/genética , Infecções por Picornaviridae/genética , Polimorfismo de Nucleotídeo Único , Mucosa Respiratória/patologia , Rhinovirus/fisiologia , Alelos , Asma/complicações , Asma/genética , Brônquios/patologia , Proteínas Relacionadas a Caderinas , Caderinas/fisiologia , Cílios/química , Células Epiteliais/química , Células Epiteliais/ultraestrutura , Predisposição Genética para Doença , Genótipo , Humanos , Proteínas de Membrana/fisiologia , Infecções por Picornaviridae/virologia , Mucosa Respiratória/virologia , Frações Subcelulares/química
2.
Respir Res ; 18(1): 84, 2017 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-28472984

RESUMO

BACKGROUND: The Rhinovirus C (RV-C), first identified in 2006, produce high symptom burdens in children and asthmatics, however, their primary target host cell in the airways remains unknown. Our primary hypotheses were that RV-C target ciliated airway epithelial cells (AECs), and that cell specificity is determined by restricted and high expression of the only known RV-C cell-entry factor, cadherin related family member 3 (CDHR3). METHODS: RV-C15 (C15) infection in differentiated human bronchial epithelial cell (HBEC) cultures was assessed using immunofluorescent and time-lapse epifluorescent imaging. Morphology of C15-infected differentiated AECs was assessed by immunohistochemistry. RESULTS: C15 produced a scattered pattern of infection, and infected cells were shed from the epithelium. The percentage of cells infected with C15 varied from 1.4 to 14.7% depending on cell culture conditions. Infected cells had increased staining for markers of ciliated cells (acetylated-alpha-tubulin [aat], p < 0.001) but not markers of goblet cells (wheat germ agglutinin or Muc5AC, p = ns). CDHR3 expression was increased on ciliated epithelial cells, but not other epithelial cells (p < 0.01). C15 infection caused a 27.4% reduction of ciliated cells expressing CDHR3 (p < 0.01). During differentiation of AECs, CDHR3 expression progressively increased and correlated with both RV-C binding and replication. CONCLUSIONS: The RV-C only replicate in ciliated AECs in vitro, leading to infected cell shedding. CDHR3 expression positively correlates with RV-C binding and replication, and is largely confined to ciliated AECs. Our data imply that factors regulating differentiation and CDHR3 production may be important determinants of RV-C illness severity.


Assuntos
Brônquios/citologia , Brônquios/virologia , Enterovirus/fisiologia , Células Epiteliais/citologia , Células Epiteliais/virologia , Internalização do Vírus , Replicação Viral/fisiologia , Células Cultivadas , Cílios/fisiologia , Cílios/ultraestrutura , Cílios/virologia , Enterovirus/ultraestrutura , Humanos , Eliminação de Partículas Virais/fisiologia
4.
J Allergy Clin Immunol ; 134(2): 332-41, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24636084

RESUMO

BACKGROUND: Epidemiologic studies provide evidence of differential virulence of rhinovirus species (RV). We recently reported that RV-A and RV-C induced more severe illnesses than RV-B, which suggests that the biology of RV-B might be different from RV-A or RV-C. OBJECTIVE: To test the hypothesis that RV-B has lower replication and induces lesser cytokine responses than RV-A or RV-C. METHODS: We cloned full-length cDNA of RV-A16, A36, B52, B72, C2, C15, and C41 from clinical samples and grew clinical isolates of RV-A7 and RV-B6 in cultured cells. Sinus epithelial cells were differentiated at the air-liquid interface. We tested for differences in viral replication in epithelial cells after infection with purified viruses (10(8) RNA copies) and measured virus load by quantitative RT-PCR. We measured lactate dehydrogenase (LDH) concentration as a marker of cellular cytotoxicity, and cytokine and/or chemokine secretion by multiplex ELISA. RESULTS: At 24 hours after infection, the virus load of RV-B (RV-B52, RV-B72, or RV-B6) in adherent cells was lower than that of RV-A or RV-C. The growth kinetics of infection indicated that RV-B types replicate more slowly. Furthermore, RV-B released less LDH than RV-A or RV-C, and induced lower levels of cytokines and chemokines such as CXCL10, even after correction for viral replication. RV-B replicates to lower levels also in primary bronchial epithelial cells. CONCLUSIONS: Our results indicate that RV-B types have lower and slower replication, and lower cellular cytotoxicity and cytokine and/or chemokine production compared with RV-A or RV-C. These characteristics may contribute to reduced severity of illnesses that has been observed with RV-B infections.


Assuntos
Brônquios/virologia , Citocinas/biossíntese , Células Epiteliais/virologia , Rhinovirus/fisiologia , Replicação Viral , Asma/complicações , Asma/imunologia , Asma/patologia , Asma/virologia , Biomarcadores/metabolismo , Brônquios/imunologia , Brônquios/patologia , Células Cultivadas , Citocinas/metabolismo , Células Epiteliais/imunologia , Células Epiteliais/patologia , Humanos , Inflamação/complicações , Inflamação/imunologia , Inflamação/patologia , Inflamação/virologia , L-Lactato Desidrogenase/metabolismo , Infecções por Picornaviridae/complicações , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Rhinovirus/isolamento & purificação , Índice de Gravidade de Doença , Especificidade da Espécie , Carga Viral
5.
Respir Res ; 14: 98, 2013 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-24219422

RESUMO

BACKGROUND: Combination therapy with budesonide and formoterol reduces exacerbations of asthma, which are closely associated with human rhinovirus (RV) infections in both children and adults. These data suggest that budesonide and formoterol inhibit virus-induced inflammatory responses of airway epithelial cells. METHODS: To test this hypothesis, bronchial epithelial (BE) cells were obtained from airway brushings of 8 subjects with moderate-to-severe allergic asthma and 9 with neither asthma nor respiratory allergies. Cultured BE cells were incubated for 24 hours with budesonide (1.77 µM), formoterol (0.1 µM), both, or neither, and then inoculated with RV-16 (5×10(6) plaque forming units [PFU]/mL). After 24 hours, viral replication (RV RNA), cytokine secretion (CXCL8, CXCL10, TNFa, IFN-ß, IL-28) and mRNA expression (CXCL8, CXCL10, TNF, IFNB1, IL-28) were analyzed. RESULTS: RV infection induced CXCL10 protein secretion and IFNB1 and IL28 mRNA expression. Drug treatments significantly inhibited secretion of CXCL10 in mock-infected, but not RV-infected, BE cells, and inhibited secretion of TNFa under both conditions. Neither budesonide nor formoterol, alone or in combination, significantly affected viral replication, nor did they inhibit RV-induced upregulation of IFNB1 and IL28 mRNA. Overall, RV replication was positively related to CXCL10 secretion and induction of IFNB1 and IL28 mRNA, but the positive relationship between RV RNA and CXCL10 secretion was stronger in normal subjects than in subjects with asthma. CONCLUSIONS: Budesonide and formoterol can inhibit BE cell inflammatory responses in vitro without interfering with viral replication or production of interferons. These effects could potentially contribute to beneficial effects of budesonide/formoterol combination therapy in preventing RV-induced asthma exacerbations.


Assuntos
Brônquios/efeitos dos fármacos , Broncodilatadores/farmacologia , Budesonida/farmacologia , Células Epiteliais/efeitos dos fármacos , Etanolaminas/farmacologia , Rhinovirus/fisiologia , Replicação Viral/efeitos dos fármacos , Adulto , Asma/etiologia , Asma/metabolismo , Asma/patologia , Brônquios/metabolismo , Brônquios/virologia , Broncodilatadores/uso terapêutico , Budesonida/uso terapêutico , Estudos de Casos e Controles , Quimiocina CXCL10/metabolismo , Citocinas/metabolismo , Quimioterapia Combinada , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Etanolaminas/uso terapêutico , Feminino , Fumarato de Formoterol , Humanos , Técnicas In Vitro , Masculino , Pessoa de Meia-Idade , Infecções por Picornaviridae/complicações , Fator de Necrose Tumoral alfa/metabolismo
6.
Mucosal Immunol ; 16(4): 386-398, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36796588

RESUMO

Rhinoviruses infect ciliated airway epithelial cells, and rhinoviruses' nonstructural proteins quickly inhibit and divert cellular processes for viral replication. However, the epithelium can mount a robust innate antiviral immune response. Therefore, we hypothesized that uninfected cells contribute significantly to the antiviral immune response in the airway epithelium. Using single-cell RNA sequencing, we demonstrate that both infected and uninfected cells upregulate antiviral genes (e.g. MX1, IFIT2, IFIH1, and OAS3) with nearly identical kinetics, whereas uninfected non-ciliated cells are the primary source of proinflammatory chemokines. Furthermore, we identified a subset of highly infectable ciliated epithelial cells with minimal interferon responses and determined that interferon responses originate from distinct subsets of ciliated cells with moderate viral replication. These findings suggest that the composition of ciliated airway epithelial cells and coordinated responses of infected and uninfected cells could determine the risk of more severe viral respiratory illnesses in children with asthma, chronic obstructive pulmonary disease, and genetically susceptible individuals.


Assuntos
Células Epiteliais , Interferons , Criança , Humanos , Células Cultivadas , Imunidade Inata , Expressão Gênica , Rhinovirus
7.
Front Cell Infect Microbiol ; 12: 1060748, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36733852

RESUMO

Rhinovirus causes many types of respiratory illnesses, ranging from minor colds to exacerbations of asthma. Moraxella catarrhalis is an opportunistic pathogen that is increased in abundance during rhinovirus illnesses and asthma exacerbations and is associated with increased severity of illness through mechanisms that are ill-defined. We used a co-infection model of human airway epithelium differentiated at the air-liquid interface to test the hypothesis that rhinovirus infection promotes M. catarrhalis adhesion and survival on the respiratory epithelium. Initial experiments showed that infection with M. catarrhalis alone did not damage the epithelium or induce cytokine production, but increased trans-epithelial electrical resistance, indicative of increased barrier function. In a co-infection model, infection with the more virulent rhinovirus-A and rhinovirus-C, but not the less virulent rhinovirus-B types, increased cell-associated M. catarrhalis. Immunofluorescent staining demonstrated that M. catarrhalis adhered to rhinovirus-infected ciliated epithelial cells and infected cells being extruded from the epithelium. Rhinovirus induced pronounced changes in gene expression and secretion of inflammatory cytokines. In contrast, M. catarrhalis caused minimal effects and did not enhance RV-induced responses. Our results indicate that rhinovirus-A or C infection increases M. catarrhalis survival and cell association while M. catarrhalis infection alone does not cause cytopathology or epithelial inflammation. Our findings suggest that rhinovirus and M. catarrhalis co-infection could promote epithelial damage and more severe illness by amplifying leukocyte inflammatory responses at the epithelial surface.


Assuntos
Asma , Coinfecção , Infecções por Enterovirus , Humanos , Moraxella catarrhalis , Rhinovirus , Coinfecção/complicações , Mucosa Respiratória , Asma/complicações , Células Epiteliais/metabolismo
8.
Cell Host Microbe ; 27(4): 614-628.e6, 2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32130954

RESUMO

Airway epithelium is the first body surface to contact inhaled irritants and report danger. Here, we report how epithelial cells recognize and respond to aeroallergen alkaline protease 1 (Alp1) of Aspergillus sp., because proteases are critical components of many allergens that provoke asthma. In a murine model, Alp1 elicits helper T (Th) cell-dependent lung eosinophilia that is initiated by the rapid response of bronchiolar club cells to Alp1. Alp1 damages bronchiolar cell junctions, which triggers a calcium flux signaled through calcineurin within club cells of the bronchioles, inciting inflammation. In two human cohorts, we link fungal sensitization and/or asthma with SNP/protein expression of the mechanosensitive calcium channel, TRPV4. TRPV4 is also necessary and sufficient for club cells to sensitize mice to Alp1. Thus, club cells detect junction damage as mechanical stress, which signals danger via TRPV4, calcium, and calcineurin to initiate allergic sensitization.


Assuntos
Aspergillus fumigatus/metabolismo , Asma/etiologia , Serina Endopeptidases/metabolismo , Canais de Cátion TRPV/metabolismo , Alérgenos/efeitos adversos , Alérgenos/metabolismo , Animais , Aspergillus fumigatus/imunologia , Bronquíolos/citologia , Calcineurina/metabolismo , Canais de Cálcio/metabolismo , Sinalização do Cálcio , Estudos de Coortes , Eosinofilia , Células Epiteliais/metabolismo , Humanos , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Serina Endopeptidases/efeitos adversos , Linfócitos T/imunologia
9.
J Immunol Methods ; 339(2): 264-9, 2008 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-18824171

RESUMO

Human rhinoviruses (HRV) are the primary etiological agents in cold infections, and represent a serious risk to individuals with chronic respiratory disease such as asthma. In order to develop treatment options for HRV infections, murine models are a crucial component in the study of infection mechanisms due to the wide array of reagents and techniques available to study murine immunology. We present here a cell culture system for studying isolated murine epithelial cell responses to HRV. Monolayers of primary mouse airway epithelial cells were maintained in a serial culture system, and the identity and purity of the cell population was confirmed via immunostaining (positive for cytokeratin, negative for vimentin). Infection of these cells with a minor group rhinovirus (HRV-1A) was evidenced by increases in viral RNA, de novo synthesis of viral proteins, and production of infectious virus. This model will be useful in experiments to define mechanisms of viral replication and host/virus interactions within airway epithelial cells.


Assuntos
Células Epiteliais/virologia , Mucosa Respiratória/virologia , Rhinovirus/crescimento & desenvolvimento , Replicação Viral , Animais , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Rhinovirus/fisiologia , Ensaio de Placa Viral/métodos , Replicação Viral/fisiologia
10.
Virus Res ; 133(2): 269-79, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18329747

RESUMO

In the late 1990s, triple reassortant H3N2 influenza A viruses emerged and spread widely within the swine population of the United States. We have shown previously that an isolate representative of this lineage of viruses, A/Swine/Minnesota/593/99 (Sw/MN), has higher infectivity and accelerated replication kinetics in pigs, compared to a human-lineage H3N2 virus isolated from a pig during the same time period, A/Swine/Ontario/00130/97 (Sw/ONT [Landolt, G.A., Karasin, A.I., Phillips, L., Olsen, C.W., 2003. Comparison of the pathogenesis of two genetically different H3N2 influenza A viruses in pigs. J. Clin. Microbiol. 41, 1936-1941]). Additional in vivo experiments using reverse genetics-generated reassortant viruses demonstrated that these phenotypes are dependent upon the HA and/or NA genes (Landolt, G.A., Karasin, A.I., Schutten, M.M., Olsen, C.W., 2006. Restricted infectivity of a human-lineage H3N2 influenza A virus in pigs is hemagglutinin and neuraminidase gene dependent. J. Clin. Microbiol. 44, 297-301). To further study the infectivity of influenza viruses for pigs, we developed a primary swine respiratory epithelial cell (SREC) culture model. In SRECs, Sw/MN infects a significantly higher number of cells compared to Sw/ONT. Using reverse genetics-generated Sw/MN x Sw/ONT reassortant viruses we demonstrate that the infectivity phenotypes of these viruses in SRECs are strongly dependent upon the HA gene. Using chimeras and point directed mutations within the HA genes, we have identified amino acids that, either alone or in combination with other amino acids, impact infectivity. In particular, amino acid 138 is the dominant factor in determining infectivity levels in SRECs.


Assuntos
Aminoácidos/genética , Células Epiteliais/virologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Vírus da Influenza A Subtipo H3N2/patogenicidade , Vírus da Influenza A/patogenicidade , Sistema Respiratório/citologia , Aminoácidos/química , Animais , Linhagem Celular , Células Cultivadas , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Humanos , Vírus da Influenza A Subtipo H3N2/genética , Vírus da Influenza A/classificação , Vírus da Influenza A/genética , Infecções por Orthomyxoviridae/veterinária , Infecções por Orthomyxoviridae/virologia , Mutação Puntual , Suínos
11.
Nat Commun ; 8(1): 1770, 2017 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-29176665

RESUMO

We inhale respiratory pathogens continuously, and the subsequent signaling events between host and microbe are complex, ultimately resulting in clearance of the microbe, stable colonization of the host, or active disease. Traditional in vitro methods are ill-equipped to study these critical events in the context of the lung microenvironment. Here we introduce a microscale organotypic model of the human bronchiole for studying pulmonary infection. By leveraging microscale techniques, the model is designed to approximate the structure of the human bronchiole, containing airway, vascular, and extracellular matrix compartments. To complement direct infection of the organotypic bronchiole, we present a clickable extension that facilitates volatile compound communication between microbial populations and the host model. Using Aspergillus fumigatus, a respiratory pathogen, we characterize the inflammatory response of the organotypic bronchiole to infection. Finally, we demonstrate multikingdom, volatile-mediated communication between the organotypic bronchiole and cultures of Aspergillus fumigatus and Pseudomonas aeruginosa.


Assuntos
Aspergillus fumigatus/metabolismo , Bronquíolos/microbiologia , Pseudomonas aeruginosa/metabolismo , Compostos Orgânicos Voláteis/metabolismo , Aspergilose/imunologia , Aspergilose/microbiologia , Aspergillus fumigatus/química , Bronquíolos/imunologia , Citocinas/imunologia , Interações Hospedeiro-Patógeno , Humanos , Pneumopatias/microbiologia , Modelos Biológicos , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/química , Compostos Orgânicos Voláteis/química
12.
PLoS One ; 9(1): e86755, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24475177

RESUMO

Vitamin D has been linked to reduced risk of viral respiratory illness. We hypothesized that vitamin D could directly reduce rhinovirus (RV) replication in airway epithelium. Primary human bronchial epithelial cells (hBEC) were treated with vitamin D, and RV replication and gene expression were evaluated by quantitative PCR. Cytokine/chemokine secretion was measured by ELISA, and transepithelial resistance (TER) was determined using a voltohmmeter. Morphology was examined using immunohistochemistry. Vitamin D supplementation had no significant effects on RV replication, but potentiated secretion of CXCL8 and CXCL10 from infected or uninfected cells. Treatment with vitamin D in the form of 1,25(OH)2D caused significant changes in cell morphology, including thickening of the cell layers (median of 46.5 µm [35.0-69.0] vs. 30 µm [24.5-34.2], p<0.01) and proliferation of cytokeratin-5-expressing cells, as demonstrated by immunohistochemical analysis. Similar effects were seen for 25(OH)D. In addition to altering morphology, higher concentrations of vitamin D significantly upregulated small proline-rich protein (SPRR1ß) expression (6.3 fold-induction, p<0.01), suggestive of squamous metaplasia. Vitamin D treatment of hBECs did not alter repair of mechanically induced wounds. Collectively, these findings indicate that vitamin D does not directly affect RV replication in airway epithelial cells, but can influence chemokine synthesis and alters the growth and differentiation of airway epithelial cells.


Assuntos
Calcitriol/farmacologia , Células Epiteliais/efeitos dos fármacos , Rhinovirus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Brônquios/efeitos dos fármacos , Brônquios/patologia , Brônquios/virologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Quimiocina CXCL10/biossíntese , Quimiocina CXCL10/metabolismo , Proteínas Ricas em Prolina do Estrato Córneo/agonistas , Proteínas Ricas em Prolina do Estrato Córneo/genética , Proteínas Ricas em Prolina do Estrato Córneo/metabolismo , Células Epiteliais/patologia , Células Epiteliais/ultraestrutura , Células Epiteliais/virologia , Expressão Gênica , Humanos , Interleucina-8/biossíntese , Interleucina-8/metabolismo , Queratina-5/agonistas , Queratina-5/genética , Queratina-5/metabolismo , Cultura Primária de Células , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/patologia , Mucosa Respiratória/virologia , Rhinovirus/fisiologia , Cicatrização/efeitos dos fármacos
13.
Cell Biol Int ; 26(1): 43-54, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11779220

RESUMO

Tropical pulmonary eosinophilia is in part caused by the hyperimmune responsiveness of the lung tissue against the antigens of degenerating microfilariae. We have previously shown that the activation of the transcription factor NF-kappaB is essential for the synthesis and release of multiple pro-inflammatory cytokines in HEp-2 human airway epithelial cells following exposure to filarial parasitic sheath proteins (FPS). Neither the antigenic component nor the receptor involved in this activation is known. Herein we provide evidence that FPS activation of NF-kappaB can be augmented by the cell surface expression of CD14. CD14 expression, however, is not sufficient to transduce FPS signals for NF-kappaB activation, since its expression in different cell types does not always furnish the capacity to respond to FPS. We also show that NF-kappaB activation by FPS treatment can be distinguished from that induced by bacterial lipolysaccharide, an agent that can also activate NF-kappaB in a CD14-dependent fashion. These observations suggest that the capacity of certain lung epithelial cells to interact with microfilarial antigens, activate NF-kappaB in a CD14-dependent manner and produce pro-inflammatory cytokines may be a contributory factor to immune responses manifested by tropical pulmonary eosinophilia.


Assuntos
Proteínas I-kappa B , Receptores de Lipopolissacarídeos/biossíntese , NF-kappa B/metabolismo , Animais , Antibacterianos/farmacologia , Western Blotting , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Endotoxinas/farmacologia , Eosinófilos , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Humanos , Pulmão/microbiologia , Camundongos , Inibidor de NF-kappaB alfa , Fosforilação , Polimixina B/farmacologia , Ligação Proteica , Transdução de Sinais , Fatores de Tempo , Transfecção
14.
Am J Respir Cell Mol Biol ; 26(5): 594-601, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11970912

RESUMO

Respiratory viruses, including rhinoviruses, infect respiratory epithelium and induce a variety of cytokines and chemokines that can initiate an inflammatory response. Cytokines, such as interferon (IFN)-gamma and tumor necrosis factor (TNF)-alpha, could enhance epithelial cell activation by inducing virus receptors. To test this hypothesis, effects of IFN-gamma or TNF-alpha on expression of intercellular adhesion molecule (ICAM)-1, rhinovirus binding, and virus-induced chemokine secretion on A549 and human bronchial epithelial cells (HBEC) were determined. The results varied with the type of cell. IFN-gamma was a stronger inducer of ICAM-1 and viral binding on HBEC, whereas TNF-alpha had greater effects on A549 cells. In addition, IFN-gamma, but not TNF-alpha, synergistically enhanced regulated on activation, normal T cells expressed and secreted (RANTES) mRNA expression and protein secretion induced by RV16 or RV49. To determine whether IFN-gamma could enhance RANTES secretion independent of effects on ICAM-1 and RV binding, HBEC were transfected with RV16 RNA in the presence or absence of IFN-gamma. RV16 RNA alone stimulated RANTES secretion, and this effect was enhanced by IFN-gamma. These results demonstrate that IFN-gamma can enhance rhinovirus-induced RANTES secretion by increasing viral binding, and through a second receptor-independent pathway. These findings suggest that IFN-gamma, by upregulating RANTES secretion, could be an important regulator of the initial immune response to rhinovirus infections.


Assuntos
Antivirais/farmacologia , Quimiocina CCL5/metabolismo , Interferon gama/farmacologia , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo , Rhinovirus/metabolismo , Células Cultivadas , Sinergismo Farmacológico , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Humanos , Molécula 1 de Adesão Intercelular/biossíntese , Ligação Proteica/efeitos dos fármacos , RNA de Cadeia Dupla/farmacologia , RNA Viral/administração & dosagem , RNA Viral/metabolismo , Mucosa Respiratória/citologia , Mucosa Respiratória/virologia , Rhinovirus/efeitos dos fármacos , Transfecção , Fator de Necrose Tumoral alfa/farmacologia , Replicação Viral/efeitos dos fármacos
15.
Am J Respir Cell Mol Biol ; 28(6): 731-7, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12600836

RESUMO

Virus-induced secretion of proinflammatory chemokines (e.g., regulated on activation, normal T cells expressed and secreted [RANTES], interleukin [IL]-8) by airway epithelial cells helps to initiate antiviral responses and airway inflammation by enhancing inflammatory cell recruitment. To define mechanisms for virus-induced chemokine secretion, monolayers of nontransformed bronchial epithelial cells were transfected or incubated with polydeoxyinosinic-deoxycytidylic acid (synthetic double-stranded [ds] RNA), rhinovirus dsRNA, or single-stranded RNA (ssRNA), and the secretion of selected chemokines was determined. Transfection or incubation with dsRNA, but not ssRNA, significantly enhanced secretion of RANTES and IL-8, but not eotaxin or macrophage inflammatory protein-1alpha. Mechanistically, dsRNA induced and activated dsRNA-dependent protein kinase (PKR), and activated nuclear factor-kappaB and p38 mitogen-activated protein kinase. Furthermore, the PKR inhibitor 2-aminopurine significantly blocked dsRNA-induced RANTES and IL-8 secretion, whereas the p38 mitogen-activated protein kinase inhibitor SB203580 suppressed dsRNA-induced IL-8, but not RANTES. These findings indicate that dsRNA selectively induce the secretion of chemokines such as IL-8 and RANTES, and implicate dsRNA-sensitive signaling proteins in this process. Moreover, these data suggest that this may be an important mechanism for the selective secretion of chemokines by viruses (e.g., rhinovirus, respiratory syncytial virus, influenza) that synthesize dsRNA during replication.


Assuntos
Brônquios/citologia , Quimiocinas/biossíntese , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , RNA de Cadeia Dupla/farmacologia , Linhagem Celular Transformada , Células Cultivadas , Quimiocina CCL11 , Quimiocina CCL5/metabolismo , Quimiocinas CC/metabolismo , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Células Epiteliais/citologia , Humanos , Imidazóis/farmacologia , Interleucina-8/metabolismo , Cinética , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Polinucleotídeos/química , Polinucleotídeos/farmacologia , Piridinas/farmacologia , RNA de Cadeia Dupla/síntese química , RNA Viral/farmacologia , Rhinovirus/genética , Transdução de Sinais , Transfecção , eIF-2 Quinase/antagonistas & inibidores , eIF-2 Quinase/efeitos dos fármacos , eIF-2 Quinase/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA