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











Base de dados
Intervalo de ano de publicação
1.
Viruses ; 15(3)2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36992456

RESUMO

Rhinoviruses (RVs) are major instigators of acute exacerbations of asthma, COPD, and other respiratory diseases. RVs are categorized into three species (RV-A, RV-B, and RV-C), which comprise more than 160 serotypes, making it difficult to develop an effective vaccine. Currently, no effective treatment for RV infection is available. Pulmonary surfactant is an extracellular complex of lipids and proteins that plays a central role in regulating innate immunity in the lung. The minor pulmonary surfactant lipids, palmitoyl-oleoyl-phosphatidylglycerol (POPG) and phosphatidylinositol (PI), are potent regulators of inflammatory processes and exert antiviral activity against respiratory syncytial virus (RSV) and influenza A viruses (IAV). In the current study, we examined the potencies of POPG and PI against rhinovirus A16 (RV-A16) in primary human airway epithelial cells (AECs) differentiated at an air-liquid interface (ALI). After AECs were infected with RV-A16, PI reduced the viral RNA copy number by 70% and downregulated (55-75%) the expression of antiviral (MDA5, IRF7, and IFN-lambda) and CXCL11 chemokine genes. In contrast, POPG only slightly decreased MDA5 (24%) and IRF7 (11%) gene expression but did not inhibit IFN-lambda gene expression or RV-A16 replication in AECs. However, both POPG and PI inhibited (50-80%) IL6 gene expression and protein secretion and CXCL11 protein secretion. PI treatment dramatically attenuated global gene expression changes induced by RV-A16 infection alone in AECs. The observed inhibitory effects were indirect and resulted mainly from the inhibition of virus replication. Cell-type enrichment analysis of viral-regulated genes opposed by PI treatment revealed the PI-inhibited viral induction of goblet cell metaplasia and the virus-induced downregulation of ciliated, club, and ionocyte cell types. Notably, the PI treatment also altered the ability of RV-A16 to regulate the expression of some phosphatidylinositol 4-kinase (PI4K); acyl-CoA-binding, domain-containing (ACBD); and low-density lipoprotein receptor (LDLR) genes that play critical roles in the formation and functioning of replication organelles (ROs) required for RV replication in host cells. These data suggest PI can be used as a potent, non-toxic, antiviral agent for RV infection prophylaxis and treatment.


Assuntos
Infecções por Enterovirus , Infecções por Picornaviridae , Surfactantes Pulmonares , Humanos , Surfactantes Pulmonares/farmacologia , Rhinovirus/genética , Células Epiteliais , Epitélio/metabolismo , Antivirais/farmacologia , Antivirais/uso terapêutico , Infecções por Enterovirus/tratamento farmacológico , Pulmão/metabolismo , Lipídeos
2.
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
3.
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
4.
Clin Exp Allergy ; 51(4): 564-573, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33471392

RESUMO

BACKGROUND: In asthma, IL-6 is a potential cause of enhanced inflammation, tissue damage and airway dysfunction. IL-6 signalling is regulated by its receptor, which is composed of two proteins, IL-6R and GP130. In addition to their membrane form, these two proteins may be found as extracellular soluble forms. The interaction of IL-6 with soluble IL-6R (sIL-6R) can trigger IL-6 trans-signalling in cells lacking IL-6R. Conversely, the soluble form of GP130 (sGP130) competes with its membrane form to inhibit IL-6 trans-signalling. OBJECTIVES: We aimed to analyse IL-6 trans-signalling proteins in the airways of subjects after an allergen challenge. METHODS: We used a model of segmental bronchoprovocation with an allergen (SBP-Ag) in human subjects with allergy. Before and 48 h after SBP-Ag, bronchoalveolar lavages (BALs) allowed for the analysis of proteins in BAL fluids (BALFs) by ELISA, and membrane proteins on the surface of BAL cells by flow cytometry. In addition, we performed RNA sequencing (RNA-seq) and used proteomic data to further inform on the expression of the IL-6R subunits by eosinophils, bronchial epithelial cells and lung fibroblasts. Finally, we measured the effect of IL-6 trans-signalling on bronchial fibroblasts, in vitro. RESULTS: IL-6, sIL-6R, sGP130 and the molar ratio of sIL-6R/sGP130 increased in the airways after SBP-Ag, suggesting the potential for enhanced IL-6 trans-signalling activity. BAL lymphocytes, monocytes and eosinophils displayed IL-6R on their surface and were all possible providers of sIL-6R, whereas GP130 was highly expressed in bronchial epithelial cells and lung fibroblasts. Finally, bronchial fibroblasts activated by IL-6 trans-signalling produced enhanced amounts of the chemokine, MCP-1 (CCL2). CONCLUSION AND CLINICAL RELEVANCE: After a bronchial allergen challenge, we found augmentation of the elements of IL-6 trans-signalling. Allergen-induced IL-6 trans-signalling activity can activate fibroblasts to produce chemokines that can further enhance inflammation and lung dysfunction.


Assuntos
Asma/metabolismo , Receptor gp130 de Citocina/metabolismo , Interleucina-6/metabolismo , Receptores de Interleucina-6/metabolismo , Alérgenos , Ambrosia , Animais , Asma/genética , Testes de Provocação Brônquica , Líquido da Lavagem Broncoalveolar/química , Quimiocina CCL2/metabolismo , Receptor gp130 de Citocina/genética , Alérgenos Animais , Feminino , Humanos , Interleucina-6/genética , Masculino , Pyroglyphidae , RNA-Seq , Receptores de Interleucina-6/genética , Hipersensibilidade Respiratória/genética , Hipersensibilidade Respiratória/metabolismo , Adulto Jovem
5.
Commun Biol ; 3(1): 678, 2020 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-33188283

RESUMO

There is a life-long relationship between rhinovirus (RV) infection and the development and clinical manifestations of asthma. In this study we demonstrate that cultured primary bronchial epithelial cells from adults with asthma (n = 9) show different transcriptional and chromatin responses to RV infection compared to those without asthma (n = 9). Both the number and magnitude of transcriptional and chromatin responses to RV were muted in cells from asthma cases compared to controls. Pathway analysis of the transcriptionally responsive genes revealed enrichments of apoptotic pathways in controls but inflammatory pathways in asthma cases. Using promoter capture Hi-C we tethered regions of RV-responsive chromatin to RV-responsive genes and showed enrichment of these regions and genes at asthma GWAS loci. Taken together, our studies indicate a delayed or prolonged inflammatory state in cells from asthma cases and highlight genes that may contribute to genetic risk for asthma.


Assuntos
Asma/metabolismo , Cromatina/metabolismo , Células Epiteliais/fisiologia , Mucosa Respiratória/citologia , Rhinovirus/fisiologia , Adulto , Asma/genética , Células Cultivadas , Humanos , Transcrição Gênica
7.
Am J Respir Cell Mol Biol ; 62(6): 783-792, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32078788

RESUMO

Polymorphism at the 17q21 gene locus and wheezing responses to rhinovirus (RV) early in childhood conspire to increase the risk of developing asthma. However, the mechanisms mediating this gene-environment interaction remain unclear. In this study, we investigated the impact of one of the 17q21-encoded genes, ORMDL3 (orosomucoid-like 3), on RV replication in human epithelial cells. ORMDL3 knockdown inhibited RV-A16 replication in HeLa, BEAS-2B, A549, and NCI-H358 epithelial cell lines and primary nasal and bronchial epithelial cells. Inhibition varied by RV species, as both minor and major group RV-A subtypes RV-B52 and RV-C2 were inhibited but not RV-C15 or RV-C41. ORMDL3 siRNA did not affect expression of the major group RV-A receptor ICAM-1 or initial internalization of RV-A16. The two major outcomes of ORMDL3 activity, SPT (serine palmitoyl-CoA transferase) inhibition and endoplasmic reticulum (ER) stress induction, were further examined: silencing ORMDL3 decreased RV-induced ER stress and IFN-ß mRNA expression. However, pharmacologic induction of ER stress and concomitant increased IFN-ß inhibited RV-A16 replication. Conversely, blockade of ER stress with tauroursodeoxycholic acid augmented replication, pointing to an alternative mechanism for the effect of ORMDL3 knockdown on RV replication. In comparison, the SPT inhibitor myriocin increased RV-A16 but not RV-C15 replication and negated the inhibitory effect of ORMDL3 knockdown. Furthermore, lipidomics analysis revealed opposing regulation of specific sphingolipid species (downstream of SPT) by myriocin and ORMDL3 siRNA, correlating with the effect of these treatments on RV replication. Together, these data revealed a requirement for ORMDL3 in supporting RV replication in epithelial cells via SPT inhibition.


Assuntos
Células Epiteliais/virologia , Proteínas de Membrana/fisiologia , Rhinovirus/fisiologia , Replicação Viral , Células A549 , Asma/etiologia , Brônquios/citologia , Células Cultivadas , Cromossomos Humanos Par 17/genética , Estresse do Retículo Endoplasmático , Ácidos Graxos Monoinsaturados/farmacologia , Predisposição Genética para Doença , Genótipo , Células HeLa , Humanos , Interferon beta/biossíntese , Interferon beta/genética , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Mucosa Nasal/citologia , Infecções por Picornaviridae/complicações , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas Recombinantes/metabolismo , Rhinovirus/genética , Serina C-Palmitoiltransferase/antagonistas & inibidores , Serina C-Palmitoiltransferase/metabolismo , Esfingolipídeos/metabolismo , Ácido Tauroquenodesoxicólico/farmacologia , Replicação Viral/efeitos dos fármacos
8.
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
9.
Am J Respir Cell Mol Biol ; 60(1): 58-67, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30156431

RESUMO

Human rhinovirus (RV), the major cause of the common cold, triggers the majority of acute airway exacerbations in patients with asthma and chronic obstructive pulmonary disease. Nitric oxide, and the related metabolite S-nitrosoglutathione, are produced in the airway epithelium via nitric oxide synthase (NOS) 2 and have been shown to function in host defense against RV infection. We hypothesized that inhibitors of the S-nitrosoglutathione-metabolizing enzyme, S-nitrosoglutathione reductase (GSNOR), might potentiate the antiviral properties of airway-derived NOS2. Using in vitro models of RV-A serotype 16 (RV-A16) and mNeonGreen-H1N1pr8 infection of human airway epithelial cells, we found that treatment with a previously characterized GSNOR inhibitor (4-[[2-[[(3-cyanophenyl)methyl]thio]-4-oxothieno-[3,2-d]pyrimidin-3(4H)-yl]methyl]-benzoic acid; referred to as C3m) decreased RV-A16 replication and expression of downstream proinflammatory and antiviral mediators (e.g., RANTES [regulated upon activation, normal T cell expressed and secreted], CXCL10, and Mx1), and increased Nrf2 (nuclear factor erythroid 2-related factor 2)-dependent genes (e.g., SQSTM1 and TrxR1). In contrast, C3m had no effect on influenza virus H1N1pr8 replication. Moreover, a structurally dissimilar GSNOR inhibitor (N6022) did not alter RV replication, suggesting that the properties of C3m may be specific to rhinovirus owing to an off-target effect. Consistent with this, C3m antiviral effects were not blocked by either NOS inhibition or GSNOR knockdown but appeared to be mediated by reduced intercellular adhesion molecule 1 transcription and increased shedding of soluble intercellular adhesion molecule 1 protein. Collectively these data show that C3m has novel antirhinoviral properties that may synergize with, but are unrelated to, its GSNOR inhibitor activity.


Assuntos
Aldeído Oxirredutases/antagonistas & inibidores , Brônquios/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Inflamação/tratamento farmacológico , Infecções por Picornaviridae/tratamento farmacológico , Rhinovirus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Benzamidas/farmacologia , Brônquios/metabolismo , Brônquios/virologia , Células Cultivadas , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Humanos , Inflamação/metabolismo , Inflamação/virologia , Óxido Nítrico Sintase Tipo II/metabolismo , Infecções por Picornaviridae/metabolismo , Infecções por Picornaviridae/virologia , Pirróis/farmacologia
10.
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
11.
J Med Virol ; 88(12): 2059-2068, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27232888

RESUMO

The clinical data on the first wheezing episodes induced by different rhinovirus (RV) species are still limited. We aimed to investigate the prevalence of RV genotypes, sensitization status, and clinical characteristics of patients having a respiratory infection caused by either different RV species or other respiratory viruses. The study enrolled 111 patients (aged 3-23 months, 79% hospitalized, 76% with RV infection) with the first wheezing episode. RV-specific sequences were identified by partial sequencing of VP4/VP2 and 5' non-coding regions with 80% success rate. The investigated clinical and laboratory variables included atopic characteristics and illness severity, parental atopic illnesses, and parental smoking. Of the study children, 56% percent had >1 atopic characteristic (atopy, eczema and/or blood eosinophil count >0.4 × 109 /L) and 23% were sensitised to allergens. RV-C was detected in 58% of RV positive samples, followed by RV-A (20%) and RV-B (1.2%). Children with RV-A and RV-C induced wheezing were older (P = 0.014) and had more atopic characteristics (P = 0.001) than those with non-RV. RV-A and RV-C illnesses had shorter duration of preadmission symptoms and required more bronchodilator use at the ward than non-RV illnesses (both P < 0.05, respectively). RV-C is the most common cause of severe early wheezing. Atopic and illness severity features are associated with children having RV-A or RV-C induced first wheezing episode rather than with children having a non-RV induced wheezing. J. Med. Virol. 88:2059-2068, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Infecções por Picornaviridae/virologia , Sons Respiratórios/etiologia , Infecções Respiratórias/virologia , Rhinovirus/classificação , Rhinovirus/genética , Feminino , Finlândia/epidemiologia , Genótipo , Humanos , Lactente , Masculino , Infecções por Picornaviridae/complicações , Infecções por Picornaviridae/epidemiologia , Reação em Cadeia da Polimerase , Prevalência , Sons Respiratórios/efeitos dos fármacos , Sons Respiratórios/fisiopatologia , Infecções Respiratórias/epidemiologia , Infecções Respiratórias/etiologia , Rhinovirus/isolamento & purificação , Análise de Sequência de DNA , Índice de Gravidade de Doença , Proteínas Virais/genética
12.
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
13.
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
14.
Virology ; 436(1): 143-9, 2013 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-23199420

RESUMO

Information about the basic biological properties of human rhinovirus-C (HRV-C) viruses is lacking due to difficulties with culturing these viruses. Our objective was to develop a cell culture system to grow HRV-C. Epithelial cells from human sinuses (HSEC) were differentiated at air-liquid interface (ALI). Differentiated cultures supported 1-2 logs growth of HRV-C15 as detected by quantitative RT-PCR. Two distinguishing features of HRVs are acid lability and optimal growth at 33-34 °C. We used this system to show that HRV-C15 is neutralized by low pH (4.5). In contrast to most HRV types, replication of HRV-C15 and HRV-C41 was similar at 34 and 37°C. The HSEC ALI provides a useful tool for quantitative studies of HRV-C replication. The ability of HRV-C to grow equally well at 34°C and 37°C may contribute to the propensity for HRV-C to cause lower airway illnesses in infants and children with asthma.


Assuntos
Células Epiteliais/virologia , Seios Paranasais/virologia , Rhinovirus/crescimento & desenvolvimento , Cultura de Vírus , Replicação Viral , Linhagem Celular , Humanos , Concentração de Íons de Hidrogênio , Seios Paranasais/citologia , Rhinovirus/classificação , Rhinovirus/metabolismo , Temperatura
15.
Virus Genes ; 35(1): 65-71, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17043759

RESUMO

A total of ten infectious bronchitis virus (IBV) isolates collected from commercial chickens in Italy in 1999 were characterized by RT-PCR and sequencing of the S1 and N genes. Phylogenetic analysis based on partial S1 gene sequences showed that five field viruses clustered together with 793/B-type strains, having 91.3-98.5% nucleotide identity within the group, and one isolate had very close sequence relationship (94.6% identity) with 624/I strain. These two IBV types have been identified in Italy previously. The other three variant isolates formed novel genotype detected recently in many countries of Western Europe. For one of these variant viruses, Italy-02, which afterwards became the prototype strain, the entire S1 gene was sequenced to confirm its originality. In contrast, phylogenetic analysis of more conserved partial N gene sequences, comprising 1-300 nucleotides, revealed different clustering. Thus, three variant IBVs of novel Italy-02 genotype, which had 96.7-99.2% S1 gene nucleotide identity with each other, belonged to three separate subgroups based on N gene sequences. 624/I-type isolate Italy-06 together with Italy-03, which was undetectable using S1 gene primers, shared 97.7% and 99.3% identity, respectively, in N gene region with vaccine strain H120. Only one of the 793/B-type isolates, Italy-10, clustered with the 793/B strain sharing 99.3% partial N gene identity, whereas the other four isolates were genetically distant from them (only 87.7-89.7% identity) and formed separate homogenous subgroup. The results demonstrated that both mutations and recombination events could contribute to the genetic diversity of the Italian isolates.


Assuntos
Variação Genética , Vírus da Bronquite Infecciosa/genética , Glicoproteínas de Membrana/genética , Proteínas do Nucleocapsídeo/genética , Filogenia , Recombinação Genética , Proteínas do Envelope Viral/genética , Animais , Embrião de Galinha , Galinhas/virologia , Vírus da Bronquite Infecciosa/isolamento & purificação , Itália , Dados de Sequência Molecular , Glicoproteína da Espícula de Coronavírus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA