Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Thorax ; 77(10): 950-959, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-34716281

RESUMO

BACKGROUND AND AIMS: The chemoattractant receptor-homologous molecule expressed on T helper type 2 cells (CRTH2) antagonist timapiprant improved lung function and asthma control in a phase 2 study, with evidence suggesting reduced exacerbations. We aimed to assess whether timapiprant attenuated or prevented asthma exacerbations induced by experimental rhinovirus (RV) infection. We furthermore hypothesised that timapiprant would dampen RV-induced type 2 inflammation and consequently improve antiviral immune responses. METHODS: Atopic patients with partially controlled asthma on maintenance inhaled corticosteroids were randomised to timapiprant (n=22) or placebo (n=22) and challenged with RV-A16 3 weeks later. The primary endpoint was the cumulative lower respiratory symptom score over the 14 days post infection. Upper respiratory symptoms, spirometry, airway hyperresponsiveness, exhaled nitric oxide, RV-A16 virus load and soluble mediators in upper and lower airways samples, and CRTH2 staining in bronchial biopsies were additionally assessed before and during RV-A16 infection. RESULTS: Six subjects discontinued the study and eight were not infected; outcomes were assessed in 16 timapiprant-treated and 14 placebo-treated, successfully infected subjects. There were no differences between treatment groups in clinical exacerbation severity including cumulative lower respiratory symptom score day 0-14 (difference 3.0 (95% CI -29.0 to 17.0), p=0.78), virus load, antiviral immune responses, or RV-A16-induced airway inflammation other than in the bronchial biopsies, where CRTH2 staining was increased during RV-A16 infection in the placebo-treated but not the timapiprant-treated group. Timapiprant had a favourable safety profile, with no deaths, serious adverse events or drug-related withdrawals. CONCLUSION: Timapiprant treatment had little impact on the clinicopathological changes induced by RV-A16 infection in partially controlled asthma.


Assuntos
Asma , Rhinovirus , Humanos , Projetos Piloto , Corticosteroides/uso terapêutico , Inflamação
2.
Allergy ; 77(1): 130-142, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34169553

RESUMO

BACKGROUND: Rhinoviruses are the predominant cause of respiratory viral infections and are strongly associated with asthma exacerbations. While humoral immunity plays an important role during virus infections, cellular aspects of this response are less well understood. Here, we investigated the antiviral response of circulating B cells upon experimental rhinovirus infection in healthy individuals and asthma patients. METHODS: We purified B cells from experimentally infected healthy individuals and patients with asthma and subjected them to total RNA-sequencing. Rhinovirus-derived RNA was measured in isolated B cells using a highly sensitive PCR. B cells were stimulated with rhinovirus in vitro to further study gene expression, expression of antiviral proteins and B-cell differentiation in response rhinovirus stimulation. Protein expression of pro-inflammatory cytokines in response to rhinovirus was assessed using a proximity extension assay. RESULTS: B cells isolated from experimentally infected subjects exhibited an antiviral gene profile linked to IFN-alpha, carried viral RNA in vivo and were transiently infected by rhinovirus in vitro. B cells rapidly differentiated into plasmablasts upon rhinovirus stimulation. While B cells lacked expression of interferons in response to rhinovirus exposure, co-stimulation with rhinovirus and IFN-alpha upregulated pro-inflammatory cytokine expression suggesting a potential new function of B cells during virus infections. Asthma patients showed extensive upregulation and dysregulation of antiviral gene expression. CONCLUSION: These findings add to the understanding of systemic effects of rhinovirus infections on B-cell responses in the periphery, show potential dysregulation in patients with asthma and might also have implications during infection with other respiratory viruses.


Assuntos
Asma , Infecções por Picornaviridae , Antivirais/uso terapêutico , Citocinas/farmacologia , Humanos , Interferons , Rhinovirus
3.
J Allergy Clin Immunol ; 147(2): 510-519.e5, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33068560

RESUMO

BACKGROUND: The mechanisms underlying altered susceptibility and propensity to severe Coronavirus disease 2019 (COVID-19) disease in at-risk groups such as patients with chronic obstructive pulmonary disease (COPD) are poorly understood. Inhaled corticosteroids (ICSs) are widely used in COPD, but the extent to which these therapies protect or expose patients to risk of severe COVID-19 is unknown. OBJECTIVE: The aim of this study was to evaluate the effect of ICSs following pulmonary expression of the SARS-CoV-2 viral entry receptor angiotensin-converting enzyme-2 (ACE2). METHODS: We evaluated the effect of ICS administration on pulmonary ACE2 expression in vitro in human airway epithelial cell cultures and in vivo in mouse models of ICS administration. Mice deficient in the type I IFN-α/ß receptor (Ifnar1-/-) and administration of exogenous IFN-ß were used to study the functional role of type-I interferon signaling in ACE2 expression. We compared sputum ACE2 expression in patients with COPD stratified according to use or nonuse of ICS. RESULTS: ICS administration attenuated ACE2 expression in mice, an effect that was reversed by exogenous IFN-ß administration, and Ifnar1-/- mice had reduced ACE2 expression, indicating that type I interferon contributes mechanistically to this effect. ICS administration attenuated expression of ACE2 in airway epithelial cell cultures from patients with COPD and in mice with elastase-induced COPD-like changes. Compared with ICS nonusers, patients with COPD who were taking ICSs also had reduced sputum expression of ACE2. CONCLUSION: ICS therapies in COPD reduce expression of the SARS-CoV-2 entry receptor ACE2. This effect may thus contribute to altered susceptibility to COVID-19 in patients with COPD.


Assuntos
Corticosteroides/administração & dosagem , Enzima de Conversão de Angiotensina 2/antagonistas & inibidores , COVID-19 , Interferon Tipo I/antagonistas & inibidores , Doença Pulmonar Obstrutiva Crônica/imunologia , SARS-CoV-2 , Administração por Inalação , Idoso , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/imunologia , Animais , Brônquios/citologia , Células Cultivadas , Suscetibilidade a Doenças , Regulação para Baixo/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Feminino , Humanos , Interferon Tipo I/imunologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Doença Pulmonar Obstrutiva Crônica/genética , Receptor de Interferon alfa e beta/genética , Serina Endopeptidases/genética
4.
J Allergy Clin Immunol ; 148(4): 1016-1029.e16, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34153372

RESUMO

BACKGROUND: Respiratory infections with rhinoviruses (RV) are strongly associated with development and exacerbations of asthma, and they pose an additional health risk for subjects with allergy. OBJECTIVE: How RV infections and chronic allergic diseases are linked and what role RV plays in the breaking of tolerance in regulatory T (Treg) cells is unknown. Therefore, this study aims to investigate the effects of RV on Treg cells. METHODS: Treg cells were isolated from subjects with asthma and controls after experimental infection with the RV-A16 (RV16) and analyzed with next-generation sequencing. Additionally, suppression assays, quantitative PCR assays, and protein quantifications were performed with Treg cells after in vitro RV16 infection. RESULTS: RV16 induced a strong antiviral response in Treg cells from subjects with asthma and controls, including the upregulation of IFI44L, MX1, ISG15, IRF7, and STAT1. In subjects with asthma, the inflammatory response was exaggerated and showed a dysregulated immune response compared with that in the controls. Furthermore, subjects with asthma failed to upregulate several immunosuppressive molecules such as CTLA4 and CD69, and they upregulated the inflammasome-related genes PYCARD and AIM2. Additionally, RV16 reduced the suppressive capacity of Treg cells from healthy subjects and subjects with asthma in vitro and increased TH2 cell-type cytokine production. CONCLUSIONS: Treg cells from healthy subjects and subjects with asthma displayed an antiviral response after RV infection and showed reduced suppressive capacity. These data suggest that Treg cell function might be altered or impaired during RV infections, which might play an important role in the association between RV and the development of asthma and asthma exacerbations.


Assuntos
Asma/imunologia , Infecções por Picornaviridae/imunologia , Rhinovirus , Linfócitos T Reguladores/imunologia , Adolescente , Adulto , Citocinas/imunologia , Feminino , Humanos , Masculino , Rhinovirus/genética , Adulto Jovem
5.
Am J Respir Cell Mol Biol ; 64(3): 344-356, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33264064

RESUMO

The interplay of type-2 inflammation and antiviral immunity underpins asthma exacerbation pathogenesis. Virus infection induces type-2 inflammation-promoting chemokines CCL17 and CCL22 in asthma; however, mechanisms regulating induction are poorly understood. By using a human rhinovirus (RV) challenge model in human airway epithelial cells in vitro and mice in vivo, we assessed mechanisms regulating CCL17 and CCL22 expression. Subjects with mild to moderate asthma and healthy volunteers were experimentally infected with RV and airway CCL17 and CCL22 protein quantified. In vitro airway epithelial cell- and mouse-RV infection models were then used to define STAT6- and NF-κB-mediated regulation of CCL17 and CCL22 expression. Following RV infection, CCL17 and CCL22 expression was higher in asthma, which differentially correlated with clinical and immunological parameters. Air-liquid interface-differentiated primary epithelial cells from donors with asthma also expressed higher levels of RV-induced CCL22. RV infection boosted type-2 cytokine-induced STAT6 activation. In epithelial cells, type-2 cytokines and STAT6 activation had differential effects on chemokine expression, increasing CCL17 and suppressing CCL22, whereas NF-κB promoted expression of both chemokines. In mice, RV infection activated pulmonary STAT6, which was required for CCL17 but not CCL22 expression. STAT6-knockout mice infected with RV expressed increased levels of NF-κB-regulated chemokines, which was associated with rapid viral clearance. Therefore, RV-induced upregulation of CCL17 and CCL22 was mediated by NF-κB activation, whereas expression was differentially regulated by STAT6. Together, these findings suggest that therapeutic targeting of type-2 STAT6 activation alone will not block all inflammatory pathways during RV infection in asthma.


Assuntos
Asma/patologia , Asma/virologia , Quimiocina CCL17/metabolismo , Quimiocina CCL22/metabolismo , Progressão da Doença , Rhinovirus/fisiologia , Fator de Transcrição STAT6/metabolismo , Células A549 , Adolescente , Adulto , Animais , Biomarcadores/metabolismo , Quimiocinas/metabolismo , Células Epiteliais/metabolismo , Feminino , Humanos , Cinética , Pulmão/patologia , Pulmão/virologia , Masculino , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Doadores de Tecidos , Adulto Jovem
6.
PLoS Pathog ; 12(9): e1005913, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27683080

RESUMO

Current understanding of adaptive immune, particularly T cell, responses to human rhinoviruses (RV) is limited. Memory T cells are thought to be of a primarily T helper 1 type, but both T helper 1 and T helper 2 memory cells have been described, and heightened T helper 2/ lessened T helper 1 responses have been associated with increased RV-induced asthma exacerbation severity. We examined the contribution of T helper 1 cells to RV-induced airways inflammation using mice deficient in the transcription factor T-Box Expressed In T Cells (Tbet), a critical controller of T helper 1 cell differentiation. Using flow cytometry we showed that Tbet deficient mice lacked the T helper 1 response of wild type mice and instead developed mixed T helper 2/T helper 17 responses to RV infection, evidenced by increased numbers of GATA binding protein 3 (GATA-3) and RAR-related orphan receptor gamma t (RORγt), and interleukin-13 and interleukin-17A expressing CD4+ T cells in the lung. Forkhead box P3 (FOXP3) and interleukin-10 expressing T cell numbers were unaffected. Tbet deficient mice also displayed deficiencies in lung Natural Killer, Natural Killer T cell and γδT cell responses, and serum neutralising antibody responses. Tbet deficient mice exhibited pronounced airways eosinophilia and mucus production in response to RV infection that, by utilising a CD4+ cell depleting antibody, were found to be T helper cell dependent. RV induction of T helper 2 and T helper 17 responses may therefore have an important role in directly driving features of allergic airways disease such as eosinophilia and mucus hypersecretion during asthma exacerbations.

8.
Lancet ; 385 Suppl 1: S88, 2015 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26312910

RESUMO

BACKGROUND: Inhaled corticosteroids are commonly used in the treatment of asthma and chronic obstructive pulmonary disease, but their effects on viral loads and anti-viral responses are poorly characterised. The aim of this study was to assess effects of inhaled fluticasone propionate on rhinovirus infection in vivo, in a mouse model. We tested the hypothesis that this treatment would reduce virus-induced airways inflammation but that the effect would be confounded by interference with anti-viral immune responses, leading to delayed viral clearance. METHODS: C57BL/6 mice were intranasally dosed with fluticasone propionate (1 mg/kg) or vehicle (dimethyl sulfoxide, control), 1 h before infection with rhinovirus 1B. We assessed bronchoalveolar lavage (BAL) inflammatory cell numbers, and measured gene expression, protein production of innate mediators, or both by quantitative RT-PCR or ELISA. We compared mice treated with fluticasone with controls at various timepoints after infection. In additional experiments, recombinant interferon (IFN) beta was administered with fluticasone and rhinovirus 1B in both groups of mice. FINDINGS: At 24 h post infection, fluticasone treatment suppressed rhinovirus induction of type I and III IFNs in the airways (for the fluticasone-treated group compared with controls: mean IFNß BAL protein 20·2 pg/mL [SD 16·7] vs 103·0 [30·9]; mean IFNλ BAL protein 102·6 pg/mL [17·4] vs 217 [44·6], p<0·001); it also impaired viral clearance, with increased lung tissue viral RNA copy numbers (4·7 × 10(5) copies [SD 1·3] vs 2·6 × 10(5) [0·8], p<0·001). Despite increasing viral loads, fluticasone inhibited rhinovirus-induced airway inflammation as evidenced by suppressed BAL neutrophil numbers in fluticasone compared with control mice (0·021 × 10(5) [0·012] vs 0·59 × (5) [0·39], p<0·001) and by suppressed lymphocyte numbers (0·092 × 10(5) [0·044] vs 0·45 × 10(5) [0·11], p<0·001). By contrast, fluticasone increased MUC5AC proteins (158·2 arbitrary units [29·9] vs 107·6 [7·1], p=0·0165) and MUC5B proteins (623·8 arbitrary units [231·9] vs 413·5 [70·5], p=0·0476) in BAL at day 7 post infection. Administration of intranasal recombinant IFN beta (10(4) units) with fluticasone and rhinovirus 1B led to upregulation of interferon-inducible cytokines OAS and CXCL10/IP-10 compared with control mice treated with fluticasone and rhinovirus alone and improved viral clearance without having any effect on suppression of inflammation by fluticasone. INTERPRETATION: Our findings suggest that fluticasone treatment suppresses rhinovirus-induced airways inflammation in vivo but also impairs anti-viral immune responses and increases viral titres, leading to mucus hypersecretion. Since asthma and chronic obstructive pulmonary disease are both associated with inherent deficient IFN responses to rhinovirus, inhaled corticosteroids might interact synergistically with disease to inhibit IFN and thus lead to increased severity of exacerbation. The clinical applicability of these findings requires confirmation in human models of disease. FUNDING: Wellcome Trust.

9.
Clin Infect Dis ; 60(10): 1528-31, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25645216

RESUMO

Rhinoviruses cause the common cold and exacerbations of asthma. Animal models of infection have identified a protective role for interleukin-18 (IL-18). Following experimental rhinovirus infection, we observed increased respiratory symptoms in healthy and asthmatic subjects with low nasal and bronchial IL-18 levels.


Assuntos
Asma/imunologia , Asma/patologia , Resfriado Comum/imunologia , Resfriado Comum/patologia , Interleucina-18/imunologia , Rhinovirus/imunologia , Adulto , Asma/virologia , Resfriado Comum/virologia , Feminino , Humanos , Interleucina-18/análise , Masculino , Pessoa de Meia-Idade , Mucosa Respiratória/química
10.
PLoS Pathog ; 9(9): e1003669, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24086140

RESUMO

Human rhinovirus (RV) infections are the principle cause of common colds and precipitate asthma and COPD exacerbations. There is currently no RV vaccine, largely due to the existence of ∼150 strains. We aimed to define highly conserved areas of the RV proteome and test their usefulness as candidate antigens for a broadly cross-reactive vaccine, using a mouse infection model. Regions of the VP0 (VP4+VP2) capsid protein were identified as having high homology across RVs. Immunization with a recombinant VP0 combined with a Th1 promoting adjuvant induced systemic, antigen specific, cross-serotype, cellular and humoral immune responses. Similar cross-reactive responses were observed in the lungs of immunized mice after infection with heterologous RV strains. Immunization enhanced the generation of heterosubtypic neutralizing antibodies and lung memory T cells, and caused more rapid virus clearance. Conserved domains of the RV capsid therefore induce cross-reactive immune responses and represent candidates for a subunit RV vaccine.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Proteínas do Capsídeo/imunologia , Pulmão/imunologia , Rhinovirus/imunologia , Animais , Asma/imunologia , Asma/virologia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/farmacologia , Resfriado Comum/genética , Resfriado Comum/imunologia , Resfriado Comum/prevenção & controle , Reações Cruzadas , Feminino , Humanos , Imunização , Pulmão/virologia , Camundongos , Doença Pulmonar Obstrutiva Crônica/imunologia , Doença Pulmonar Obstrutiva Crônica/virologia , Rhinovirus/genética , Vacinas Virais
11.
PLoS Pathog ; 9(8): e1003520, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23935498

RESUMO

Human rhinoviruses (HRV) cause the majority of common colds and acute exacerbations of asthma and chronic obstructive pulmonary disease (COPD). Effective therapies are urgently needed, but no licensed treatments or vaccines currently exist. Of the 100 identified serotypes, ∼90% bind domain 1 of human intercellular adhesion molecule-1 (ICAM-1) as their cellular receptor, making this an attractive target for development of therapies; however, ICAM-1 domain 1 is also required for host defence and regulation of cell trafficking, principally via its major ligand LFA-1. Using a mouse anti-human ICAM-1 antibody (14C11) that specifically binds domain 1 of human ICAM-1, we show that 14C11 administered topically or systemically prevented entry of two major groups of rhinoviruses, HRV16 and HRV14, and reduced cellular inflammation, pro-inflammatory cytokine induction and virus load in vivo. 14C11 also reduced cellular inflammation and Th2 cytokine/chemokine production in a model of major group HRV-induced asthma exacerbation. Interestingly, 14C11 did not prevent cell adhesion via human ICAM-1/LFA-1 interactions in vitro, suggesting the epitope targeted by 14C11 was specific for viral entry. Thus a human ICAM-1 domain-1-specific antibody can prevent major group HRV entry and induction of airway inflammation in vivo.


Assuntos
Anticorpos Monoclonais Murinos/farmacologia , Imunoglobulina G/farmacologia , Molécula 1 de Adesão Intercelular/imunologia , Infecções por Picornaviridae/imunologia , Pneumonia Viral/imunologia , Rhinovirus/imunologia , Internalização do Vírus/efeitos dos fármacos , Animais , Anticorpos Monoclonais Murinos/imunologia , Quimiocinas/genética , Quimiocinas/imunologia , Células HeLa , Humanos , Imunoglobulina G/imunologia , Molécula 1 de Adesão Intercelular/genética , Células Jurkat , Antígeno-1 Associado à Função Linfocitária/genética , Antígeno-1 Associado à Função Linfocitária/imunologia , Camundongos , Camundongos Transgênicos , Infecções por Picornaviridae/tratamento farmacológico , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/patologia , Pneumonia Viral/dietoterapia , Pneumonia Viral/genética , Pneumonia Viral/patologia , Células Th2/imunologia
12.
Clin Sci (Lond) ; 129(3): 245-58, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25783022

RESUMO

Viral exacerbations of chronic obstructive pulmonary disease (COPD), commonly caused by rhinovirus (RV) infections, are poorly controlled by current therapies. This is due to a lack of understanding of the underlying immunopathological mechanisms. Human studies have identified a number of key immune responses that are associated with RV-induced exacerbations including neutrophilic inflammation, expression of inflammatory cytokines and deficiencies in innate anti-viral interferon. Animal models of COPD exacerbation are required to determine the contribution of these responses to disease pathogenesis. We aimed to develop a short-term mouse model that reproduced the hallmark features of RV-induced exacerbation of COPD. Evaluation of complex protocols involving multiple dose elastase and lipopolysaccharide (LPS) administration combined with RV1B infection showed suppression rather than enhancement of inflammatory parameters compared with control mice infected with RV1B alone. Therefore, these approaches did not accurately model the enhanced inflammation associated with RV infection in patients with COPD compared with healthy subjects. In contrast, a single elastase treatment followed by RV infection led to heightened airway neutrophilic and lymphocytic inflammation, increased expression of tumour necrosis factor (TNF)-α, C-X-C motif chemokine 10 (CXCL10)/IP-10 (interferon γ-induced protein 10) and CCL5 [chemokine (C-C motif) ligand 5]/RANTES (regulated on activation, normal T-cell expressed and secreted), mucus hypersecretion and preliminary evidence for increased airway hyper-responsiveness compared with mice treated with elastase or RV infection alone. In summary, we have developed a new mouse model of RV-induced COPD exacerbation that mimics many of the inflammatory features of human disease. This model, in conjunction with human models of disease, will provide an essential tool for studying disease mechanisms and allow testing of novel therapies with potential to be translated into clinical practice.


Assuntos
Asma/imunologia , Infecções por Picornaviridae/imunologia , Doença Pulmonar Obstrutiva Crônica/virologia , Rhinovirus/imunologia , Animais , Asma/virologia , Quimiocinas/imunologia , Modelos Animais de Doenças , Feminino , Lipopolissacarídeos/imunologia , Camundongos Endogâmicos C57BL , Doença Pulmonar Obstrutiva Crônica/imunologia , Fatores de Tempo
13.
Respirology ; 16(2): 367-77, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21199160

RESUMO

BACKGROUND AND OBJECTIVE: A hallmark of asthma is airway remodelling, which includes increased deposition of extracellular matrix (ECM) protein. Viral infections may promote the development of asthma and are the most common causes of asthma exacerbations. We evaluated whether rhinovirus (RV) infection induces airway remodelling, as assessed by ECM deposition. METHODS: Primary human bronchial epithelial cells and lung parenchymal fibroblasts were infected with RV-2 or RV-16, or treated with RV-16 RNA, imiquimod (Toll-like receptor (TLR) 7/8 agonist) or polyinosinic : polycytidylic acid (poly I : C) (activator of TLR 3, retinoic-acid-inducible protein I and melanoma-differentiated-associated gene 5). Changes in ECM proteins and their transcription were measured by ELISA and quantitative real-time PCR. In addition, gene expression for ECM proteins was assessed in a mouse model of RV infection. RESULTS: RV infection increased deposition of the ECM protein, perlecan, by human bronchial epithelial cells, and collagen V and matrix-bound vascular endothelial growth factor were increased in both human bronchial epithelial cell and fibroblast cultures. Purified RV-16 RNA, poly I : C and imiquimod induced similar increases in ECM deposition to those observed with RV-infected fibroblasts. However, only poly I : C induced ECM deposition by bronchial epithelial cells, suggesting that RV-induced ECM deposition is mediated through TLR. Furthermore, gene expression for fibronectin and collagen I was increased in lung homogenates of mice infected with RV-1b. CONCLUSIONS: RV infection and TLR ligands promote ECM deposition in isolated cell systems and RV induces ECM gene expression in vivo, thus demonstrating that RV has the potential to contribute to remodelling of the airways through induction of ECM deposition.


Assuntos
Remodelação das Vias Aéreas , Asma/virologia , Brônquios/virologia , Infecções por Picornaviridae/virologia , Aminoquinolinas/farmacologia , Animais , Asma/metabolismo , Brônquios/metabolismo , Células Cultivadas , Colágeno Tipo V/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/virologia , Proteínas da Matriz Extracelular/metabolismo , Feminino , Humanos , Imiquimode , Camundongos , Infecções por Picornaviridae/metabolismo , Poli I-C/farmacologia , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas
14.
Vaccine ; 37(21): 2805-2813, 2019 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-31003914

RESUMO

Human rhinovirus (RV) infections are the principle cause of common colds and precipitate asthma and chronic obstructive pulmonary disease (COPD) exacerbations. Currently there is no vaccine for RV which is largely due to the existence of ∼160 serotypes/strains. We demonstrated previously that immunising mice with highly conserved VP4 and VP2 regions of the RV polyprotein (RV-A16 VP0) generated cross-reactive immunity to RV in vivo. The current study investigated and mapped the epitopes of RV-A16 VP0 that are targets for antibodies in serum samples from VP0 immunisation and RV challenge studies in mice. Recombinant capsid proteins, peptide pools and individual peptides spanning the immunogen sequence (RV-A16 VP0) were assessed for IgG binding sites to identify epitopes. We found that peptide pools covering the C-terminus of VP4, the N-terminus of VP2 and the neutralising NIm-II site within VP2 were bound by serum IgG from immunised mice. The NIm-II site peptide pool blocked IgG binding to the immunogen RV-A16 VP0 and individual peptides within the pool binding IgG were further mapped. Thus, we have identified immunodominant epitopes of RV vaccine candidate RV-A16 VP0, noting that strong IgG binding antibodies were observed that target a key neutralising epitope that is highly variable amongst RV serotypes.


Assuntos
Rhinovirus/imunologia , Proteínas Virais/imunologia , Animais , Anticorpos Antivirais/imunologia , Proteínas do Capsídeo/química , Proteínas do Capsídeo/imunologia , Reações Cruzadas/imunologia , Ensaio de Imunoadsorção Enzimática , Mapeamento de Epitopos , Humanos , Epitopos Imunodominantes/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/prevenção & controle , Rhinovirus/patogenicidade , Proteínas Virais/química , Vacinas Virais/uso terapêutico
15.
Sci Transl Med ; 11(507)2019 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-31462509

RESUMO

Bacterial infection commonly complicates inflammatory airway diseases such as chronic obstructive pulmonary disease (COPD). The mechanisms of increased infection susceptibility and how use of the commonly prescribed therapy inhaled corticosteroids (ICS) accentuates pneumonia risk in COPD are poorly understood. Here, using analysis of samples from patients with COPD, we show that ICS use is associated with lung microbiota disruption leading to proliferation of streptococcal genera, an effect that could be recapitulated in ICS-treated mice. To study mechanisms underlying this effect, we used cellular and mouse models of streptococcal expansion with Streptococcus pneumoniae, an important pathogen in COPD, to demonstrate that ICS impairs pulmonary clearance of bacteria through suppression of the antimicrobial peptide cathelicidin. ICS impairment of pulmonary immunity was dependent on suppression of cathelicidin because ICS had no effect on bacterial loads in mice lacking cathelicidin (Camp -/-) and exogenous cathelicidin prevented ICS-mediated expansion of streptococci within the microbiota and improved bacterial clearance. Suppression of pulmonary immunity by ICS was mediated by augmentation of the protease cathepsin D. Collectively, these data suggest a central role for cathepsin D/cathelicidin in the suppression of antibacterial host defense by ICS in COPD. Therapeutic restoration of cathelicidin to boost antibacterial immunity and beneficially modulate the lung microbiota might be an effective strategy in COPD.


Assuntos
Corticosteroides/farmacologia , Peptídeos Catiônicos Antimicrobianos/metabolismo , Disbiose/metabolismo , Disbiose/microbiologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/microbiologia , Corticosteroides/administração & dosagem , Idoso , Animais , Peptídeos Catiônicos Antimicrobianos/farmacologia , Infecções Bacterianas/metabolismo , Infecções Bacterianas/microbiologia , Feminino , Fluticasona/farmacologia , Humanos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/microbiologia , Masculino , Camundongos , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/patogenicidade , Catelicidinas
16.
Nat Commun ; 9(1): 2229, 2018 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-29884817

RESUMO

Inhaled corticosteroids (ICS) have limited efficacy in reducing chronic obstructive pulmonary disease (COPD) exacerbations and increase pneumonia risk, through unknown mechanisms. Rhinoviruses precipitate most exacerbations and increase susceptibility to secondary bacterial infections. Here, we show that the ICS fluticasone propionate (FP) impairs innate and acquired antiviral immune responses leading to delayed virus clearance and previously unrecognised adverse effects of enhanced mucus, impaired antimicrobial peptide secretion and increased pulmonary bacterial load during virus-induced exacerbations. Exogenous interferon-ß reverses these effects. FP suppression of interferon may occur through inhibition of TLR3- and RIG-I virus-sensing pathways. Mice deficient in the type I interferon-α/ß receptor (IFNAR1-/-) have suppressed antimicrobial peptide and enhanced mucin responses to rhinovirus infection. This study identifies type I interferon as a central regulator of antibacterial immunity and mucus production. Suppression of interferon by ICS during virus-induced COPD exacerbations likely mediates pneumonia risk and raises suggestion that inhaled interferon-ß therapy may protect.


Assuntos
Corticosteroides/farmacologia , Carga Bacteriana/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Muco/efeitos dos fármacos , Doença Pulmonar Obstrutiva Crônica/prevenção & controle , Rhinovirus/efeitos dos fármacos , Administração por Inalação , Corticosteroides/administração & dosagem , Corticosteroides/imunologia , Animais , Infecções Bacterianas/microbiologia , Infecções Bacterianas/prevenção & controle , Linhagem Celular , Fluticasona/administração & dosagem , Fluticasona/imunologia , Fluticasona/farmacologia , Humanos , Pulmão/efeitos dos fármacos , Pulmão/microbiologia , Pulmão/virologia , Camundongos Knockout , Muco/microbiologia , Muco/virologia , Infecções por Picornaviridae/prevenção & controle , Infecções por Picornaviridae/virologia , Doença Pulmonar Obstrutiva Crônica/microbiologia , Doença Pulmonar Obstrutiva Crônica/virologia , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Rhinovirus/imunologia , Rhinovirus/fisiologia
17.
Curr Opin Virol ; 11: 83-8, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25829255

RESUMO

A great burden of disease is attributable to human rhinovirus (HRV) infections which are the major cause of the common cold, exacerbations of both asthma and chronic obstructive pulmonary disease (COPD), and are associated with asthma development. Despite this there is currently no vaccine for HRV. The first vaccine studies showed some promise in terms of serotype-specific protection against cold symptoms, but antigenic heterogeneity amongst the >150 HRVs has been regarded as a major barrier to effective vaccine development and has resulted in little progress over 50 years. Here we review those vaccine studies conducted to date, discuss the difficulties posed by antigenic heterogeneity and describe some recent advances in generating cross-reactive antibodies and T cell responses using peptide immunogens.


Assuntos
Imunidade Heteróloga , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/prevenção & controle , Rhinovirus/imunologia , Vacinas Virais/imunologia , Vacinas Virais/isolamento & purificação , Descoberta de Drogas/métodos , Descoberta de Drogas/tendências , Humanos , Infecções por Picornaviridae/virologia , Sorogrupo
19.
EMBO Mol Med ; 4(12): 1244-60, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23165884

RESUMO

The importance of NF-κB activation and deficient anti-viral interferon induction in the pathogenesis of rhinovirus-induced asthma exacerbations is poorly understood. We provide the first in vivo evidence in man and mouse that rhinovirus infection enhanced bronchial epithelial cell NF-κB p65 nuclear expression, NF-κB p65 DNA binding in lung tissue and NF-κB-regulated airway inflammation. In vitro inhibition of NF-κB reduced rhinovirus-induced pro-inflammatory cytokines but did not affect type I/III interferon induction. Rhinovirus-infected p65-deficient mice exhibited reduced neutrophilic inflammation, yet interferon induction, antiviral responses and virus loads were unaffected, indicating that NF-κB p65 is required for pro-inflammatory responses, but redundant in interferon induction by rhinoviruses in vivo. Conversely, IFNAR1(-/-) mice exhibited enhanced neutrophilic inflammation with impaired antiviral immunity and increased rhinovirus replication, demonstrating that interferon signalling was critical to antiviral immunity. We thus provide new mechanistic insights into rhinovirus infection and demonstrate the therapeutic potential of targeting NF-κB p65 (to suppress inflammation but preserve anti-viral immunity) and type I IFN signalling (to enhance deficient anti-viral immunity) to treat rhinovirus-induced exacerbations of airway diseases.


Assuntos
Imunidade Inata , Interferon Tipo I/metabolismo , NF-kappa B/metabolismo , Fosfoproteínas/genética , Rhinovirus/fisiologia , Animais , Asma/imunologia , Asma/metabolismo , Asma/virologia , Células Cultivadas , Proteínas do Citoesqueleto , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Feminino , Células HeLa , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Interferon Tipo I/imunologia , Pulmão/imunologia , Pulmão/metabolismo , Camundongos , Proteínas dos Microfilamentos , Fosfoproteínas/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Rhinovirus/imunologia , Rhinovirus/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA