Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 200
Filtrar
1.
J Allergy Clin Immunol ; 151(6): 1577-1584.e4, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36708816

RESUMEN

BACKGROUND: Rhinoviruses (RVs) are the most common trigger for asthma exacerbations, and there are currently no targeted therapies for viral-induced asthma exacerbations. RV infection causes neutrophilic inflammation, which is often resistant to effects of glucocorticoids. IL-1 receptor antagonist (IL-1RA) treatment reduces neutrophilic inflammation in humans challenged with inhaled endotoxin and thus may have therapeutic potential for RV-induced asthma exacerbations. OBJECTIVE: We sought to test the hypothesis that IL-1RA treatment of airway epithelium reduces RV-mediated proinflammatory cytokine production, which is important for neutrophil recruitment. METHODS: Human bronchial epithelial cells from deceased donors without prior pulmonary disease were cultured at air-liquid interface and treated with IL-13 to approximate an asthmatic inflammatory milieu. Human bronchial epithelial cells were infected with human RV-16 with or without IL-1RA treatment. RESULTS: RV infection promoted the release of IL-1α and the neutrophil-attractant cytokines IL-6, IL-8, and CXCL10. Proinflammatory cytokine secretion was significantly reduced by IL-1RA treatment without significant change in IFN-ß release or RV titer. In addition, IL-1RA reduced MUC5B expression after RV infection without impacting MUC5AC. CONCLUSIONS: These data suggest that IL-1RA treatment significantly reduced proinflammatory cytokines while preserving the antiviral response. These results provide evidence for further investigation of IL-1RA as a novel targeted therapy against neutrophil-attractant cytokine release in RV-induced airway inflammatory responses.


Asunto(s)
Asma , Infecciones por Enterovirus , Infecciones por Picornaviridae , Humanos , Rhinovirus/fisiología , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Receptores de Interleucina-1 , Asma/tratamiento farmacológico , Citocinas/metabolismo , Epitelio/metabolismo , Células Epiteliales/metabolismo , Inflamación/tratamiento farmacológico , Infecciones por Picornaviridae/tratamiento farmacológico
2.
Curr Opin Pediatr ; 35(3): 380-389, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36876331

RESUMEN

PURPOSE OF REVIEW: To review the epidemiology, clinical manifestations, and treatment strategies of nonpolio enterovirus and parechovirus (PeV) infections, and identify research gaps. RECENT FINDINGS: There is currently no approved antiviral agent for enterovirus or PeV infections, although pocapavir may be provided on a compassionate basis. Elucidation of the structure and functional features of enterovirus and PeV may lead to novel therapeutic strategies, including vaccine development. SUMMARY: Nonpolio human enterovirus and PeV are common childhood infections that are most severe among neonates and young infants. Although most infections are asymptomatic, severe disease resulting in substantial morbidity and mortality occurs worldwide and has been associated with local outbreaks. Long-term sequelae are not well understood but have been reported following neonatal infection of the central nervous system. The lack of antiviral treatment and effective vaccines highlight important knowledge gaps. Active surveillance ultimately may inform preventive strategies.


Asunto(s)
Infecciones por Enterovirus , Enterovirus , Parechovirus , Infecciones por Picornaviridae , Recién Nacido , Lactante , Humanos , Niño , Parechovirus/genética , Infecciones por Enterovirus/diagnóstico , Infecciones por Enterovirus/tratamiento farmacológico , Infecciones por Enterovirus/epidemiología , Antivirales/uso terapéutico , Brotes de Enfermedades/prevención & control , Infecciones por Picornaviridae/diagnóstico , Infecciones por Picornaviridae/tratamiento farmacológico , Infecciones por Picornaviridae/epidemiología
3.
J Med Virol ; 94(8): 3570-3580, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35474513

RESUMEN

Cell pyroptosis has received increased attention due to the associations between innate immunity and disease, and it has become a major focal point recently due to in-depth studies of cancer. With increased research on pyroptosis, scientists have discovered that it has an essential role in viral infections, especially in the occurrence and development of some picornavirus infections. Many picornaviruses, including Coxsackievirus, a71 enterovirus, human rhinovirus, encephalomyocarditis virus, and foot-and-mouth disease virus induce pyroptosis to varying degrees. This review summarized the mechanisms by which these viruses induce cell pyroptosis, which can be an effective defense against pathogen infection. However, excessive inflammasome activation or pyroptosis also can damage the host's health or aggravate disease progression. Careful approaches that acknowledge this dual effect will aid in the exploration of picornavirus infections and the mechanisms that produce the inflammatory response. This information will promote the development of drugs that can inhibit cell pyroptosis and provide new avenues for future clinical treatment.


Asunto(s)
Enterovirus , Infecciones por Picornaviridae , Picornaviridae , Virosis , Animales , Humanos , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Infecciones por Picornaviridae/tratamiento farmacológico , Piroptosis , Replicación Viral
4.
Inflamm Res ; 71(3): 357-368, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35107605

RESUMEN

OBJECTIVE: The maintenance of tight junction integrity contributes significantly to epithelial barrier function. If barrier function is destroyed, cell permeability increases and the movement of pathogens is promoted, further increasing the susceptibility to secondary infection. Here, we examined the protective effects of wogonin on rhinovirus (RV)-induced tight junction disruption. Additionally, we examined the signaling molecules responsible for anti-inflammatory activities in human nasal epithelial (HNE) cells. METHODS AND RESULTS: Primary HNE cells grown at an air-liquid interface and RPMI 2650 cells were infected apically with RV. Incubation with RV resulted in disruption of tight junction proteins (ZO-1, E-cadherin, claudin-1, and occludin) in the HNE cells. Cell viability of wogonin-treated HNE cells was measured using the MTT assay. Pretreatment with wogonin decreased RV-induced disruption of tight junctions in HNE cells. Furthermore, wogonin significantly decreased RV-induced phosphorylation of Akt/NF-κB and ERK1/2. Additionally, RV-induced generation of reactive oxygen species and RV-induced up-regulation of the production of inflammatory cytokines IL-8 and IL-6 were diminished by wogonin in HNE cells. CONCLUSION: Wogonin inhibits HRV-induced tight junction disruption via the suppression of inflammatory responses and phosphorylation of Akt/NF-κB and ERK1/2 in HNE cells. These finds will facilitate the development of novel therapeutic strategies.


Asunto(s)
Flavanonas , Proteína Quinasa 3 Activada por Mitógenos , FN-kappa B , Proteínas Proto-Oncogénicas c-akt , Rhinovirus , Uniones Estrechas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/virología , Flavanonas/farmacología , Humanos , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , FN-kappa B/metabolismo , Fosforilación/efectos de los fármacos , Infecciones por Picornaviridae/tratamiento farmacológico , Infecciones por Picornaviridae/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Rhinovirus/metabolismo , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Uniones Estrechas/patología
5.
Proc Natl Acad Sci U S A ; 116(38): 19109-19115, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31462495

RESUMEN

Viral inhibitors, such as pleconaril and vapendavir, target conserved regions in the capsids of rhinoviruses (RVs) and enteroviruses (EVs) by binding to a hydrophobic pocket in viral capsid protein 1 (VP1). In resistant RVs and EVs, bulky residues in this pocket prevent their binding. However, recently developed pyrazolopyrimidines inhibit pleconaril-resistant RVs and EVs, and computational modeling has suggested that they also bind to the hydrophobic pocket in VP1. We studied the mechanism of inhibition of pleconaril-resistant RVs using RV-B5 (1 of the 7 naturally pleconaril-resistant rhinoviruses) and OBR-5-340, a bioavailable pyrazolopyrimidine with proven in vivo activity, and determined the 3D-structure of the protein-ligand complex to 3.6 Å with cryoelectron microscopy. Our data indicate that, similar to other capsid binders, OBR-5-340 induces thermostability and inhibits viral adsorption and uncoating. However, we found that OBR-5-340 attaches closer to the entrance of the pocket than most other capsid binders, whose viral complexes have been studied so far, showing only marginal overlaps of the attachment sites. Comparing the experimentally determined 3D structure with the control, RV-B5 incubated with solvent only and determined to 3.2 Å, revealed no gross conformational changes upon OBR-5-340 binding. The pocket of the naturally OBR-5-340-resistant RV-A89 likewise incubated with OBR-5-340 and solved to 2.9 Å was empty. Pyrazolopyrimidines have a rigid molecular scaffold and may thus be less affected by a loss of entropy upon binding. They interact with less-conserved regions than known capsid binders. Overall, pyrazolopyrimidines could be more suitable for the development of new, broadly active inhibitors.


Asunto(s)
Antivirales/metabolismo , Cápside/metabolismo , Microscopía por Crioelectrón/métodos , Farmacorresistencia Viral , Oxadiazoles/farmacología , Rhinovirus/metabolismo , Proteínas Virales/química , Antivirales/farmacología , Sitios de Unión , Cápside/efectos de los fármacos , Cápside/ultraestructura , Células HeLa , Humanos , Modelos Moleculares , Estructura Molecular , Oxazoles , Infecciones por Picornaviridae/tratamiento farmacológico , Infecciones por Picornaviridae/metabolismo , Infecciones por Picornaviridae/virología , Unión Proteica , Conformación Proteica , Rhinovirus/efectos de los fármacos , Rhinovirus/ultraestructura , Relación Estructura-Actividad , Proteínas Virales/genética , Proteínas Virales/metabolismo
6.
Am J Respir Cell Mol Biol ; 65(5): 544-554, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34181859

RESUMEN

Human rhinovirus (RV) is a major risk factor for chronic obstructive pulmonary disease (COPD) and asthma exacerbations. The exploration of RV pathogenesis has been hampered by a lack of disease-relevant model systems. We performed a detailed characterization of host responses to RV infection in human lung tissue ex vivo and investigated whether these responses are disease relevant for patients with COPD and asthma. In addition, impact of the viral replication inhibitor rupintrivir was evaluated. Human precision-cut lung slices (PCLS) were infected with RV1B with or without rupintrivir. At Days 1 and 3 after infection, RV tissue localization, tissue viability, and viral load were determined. To characterize host responses to infection, mediator and whole genome analyses were performed. RV successfully replicated in PCLS airway epithelial cells and induced both antiviral and proinflammatory cytokines such as IFNα2a, CXCL10, CXCL11, IFN-γ, TNFα, and CCL5. Genomic analyses revealed that RV not only induced antiviral immune responses but also triggered changes in epithelial cell-associated pathways. Strikingly, the RV response in PCLS was reflective of gene expression changes described in patients with COPD and asthma. Although RV-induced host immune responses were abrogated by rupintrivir, RV-triggered epithelial processes were largely refractory to antiviral treatment. Detailed analysis of RV-infected human PCLS and comparison with gene signatures of patients with COPD and asthma revealed that the human RV PCLS model represents disease-relevant biological mechanisms that can be partially inhibited by a well-known antiviral compound and provide an outstanding opportunity to evaluate novel therapeutics.


Asunto(s)
Asma/genética , Interacciones Huésped-Patógeno/genética , Pulmón/virología , Infecciones por Picornaviridae/genética , Enfermedad Pulmonar Obstructiva Crónica/genética , Anciano , Antivirales/farmacología , Asma/patología , Bronquios/patología , Bronquios/fisiología , Células Epiteliales/patología , Células Epiteliales/virología , Femenino , Perfilación de la Expresión Génica , Genoma Humano , Humanos , Isoxazoles/farmacología , Pulmón/fisiología , Masculino , Persona de Mediana Edad , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , Infecciones por Picornaviridae/tratamiento farmacológico , Infecciones por Picornaviridae/patología , Enfermedad Pulmonar Obstructiva Crónica/patología , Pirrolidinonas/farmacología , Rhinovirus/patogenicidad , Valina/análogos & derivados , Valina/farmacología
7.
Proc Natl Acad Sci U S A ; 115(30): E7158-E7165, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-29987044

RESUMEN

Rhinoviruses (RVs) are responsible for the majority of upper airway infections; despite their high prevalence and the resulting economic burden, effective treatment is lacking. We report here that RV induces metabolic alterations in host cells, which offer an efficient target for antiviral intervention. We show that RV-infected cells rapidly up-regulate glucose uptake in a PI3K-dependent manner. In parallel, infected cells enhance the expression of the PI3K-regulated glucose transporter GLUT1. In-depth metabolomic analysis of RV-infected cells revealed a critical role of glucose mobilization from extracellular and intracellular pools via glycogenolysis for viral replication. Infection resulted in a highly anabolic state, including enhanced nucleotide synthesis and lipogenesis. Consistently, we observed that glucose deprivation from medium and via glycolysis inhibition by 2-deoxyglucose (2-DG) potently impairs viral replication. Metabolomic analysis showed that 2-DG specifically reverts the RV-induced anabolic reprogramming. In addition, treatment with 2-DG inhibited RV infection and inflammation in a murine model. Thus, we demonstrate that the specific metabolic fingerprint of RV infection can be used to identify new targets for therapeutic intervention.


Asunto(s)
Infecciones por Picornaviridae/metabolismo , Rhinovirus/fisiología , Replicación Viral/fisiología , Animales , Desoxiglucosa/farmacología , Femenino , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Lipogénesis/efectos de los fármacos , Lipogénesis/genética , Ratones , Nucleótidos/biosíntesis , Nucleótidos/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Infecciones por Picornaviridae/tratamiento farmacológico , Infecciones por Picornaviridae/genética , Infecciones por Picornaviridae/patología , Replicación Viral/efectos de los fármacos
8.
J Allergy Clin Immunol ; 146(3): 545-554, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32018030

RESUMEN

BACKGROUND: Rhinovirus frequently causes asthma exacerbations among children and young adults who are allergic. The interaction between allergen and rhinovirus-induced symptoms and inflammation over time is unclear. OBJECTIVE: Our aim was to compare the response to an experimental inoculation with rhinovirus-16 in allergic asthmatics with the response in healthy controls and to evaluate the effects of administrating omalizumab before and during the infection. METHODS: Two clinical trials were run in parallel. In one of these trials, the response to an experimental inoculation with rhinovirus-16 among asthmatics with high levels of total IgE was compared to the response in healthy controls. The other trial compared the effects of administering omalizumab versus placebo to asthmatics in a randomized, double-blind placebo-controlled investigation. The primary outcome for both trials compared lower respiratory tract symptoms (LRTSs) between study groups over the first 4 days of infection. RESULTS: Frequent comparisons of symptoms, lung function, and blood eosinophil counts revealed differences that were more pronounced among allergic asthmatics than among controls by days 2 and 3 after virus inoculation. Additionally, an augmentation of upper respiratory tract symptom scores and LRTS scores occurred among the atopic asthmatics versus the controls during the resolution of symptoms (P < .01 for upper respiratory symptom tract scores and P < .001 for LRTS scores). The beneficial effects of administering omalizumab on reducing LRTSs and improving lung function were strongest over the first 4 days. CONCLUSIONS: LRTSs and blood eosinophil counts were augmented and lung function was reduced among allergic asthmatics early after rhinovirus inoculation but increased late in the infection during symptom resolution. The effect of administering omalizumab on the response to rhinovirus was most pronounced during the early/innate phase of the infection.


Asunto(s)
Antialérgicos/uso terapéutico , Asma/inmunología , Inmunoglobulina E/metabolismo , Omalizumab/uso terapéutico , Infecciones por Picornaviridae/inmunología , Sistema Respiratorio/patología , Rhinovirus/fisiología , Adulto , Asma/tratamiento farmacológico , Método Doble Ciego , Femenino , Humanos , Inmunoglobulina E/inmunología , Masculino , Infecciones por Picornaviridae/tratamiento farmacológico , Efecto Placebo , Pruebas de Función Respiratoria , Sistema Respiratorio/virología , Adulto Joven
9.
Paediatr Respir Rev ; 34: 53-58, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31054799

RESUMEN

Although recent guidelines recommend a minimalist approach to bronchiolitis, there are several issues with this posture. First, there are concerns about the definition of the disease, the quality of the guidelines, the method of administration of bronchodilators, and the availability of tools to evaluate the response to therapies. Second, for decades it has been assumed that all cases of viral bronchiolitis are the same, but recent evidence has shown that this is not the case. Distinct bronchiolitis phenotypes have been described, with heterogeneity in clinical presentation, molecular immune signatures and clinically relevant outcomes such as respiratory failure and recurrent wheezing. New research is critically needed to refine viral bronchiolitis phenotyping at the molecular and clinical levels as well as to define phenotype-specific responses to different therapeutic options.


Asunto(s)
Bronquiolitis Viral/tratamiento farmacológico , Bronquiolitis Viral/fisiopatología , Broncodilatadores/uso terapéutico , Ruidos Respiratorios/fisiopatología , Péptidos Catiónicos Antimicrobianos/inmunología , Bronquiolitis Viral/inmunología , Bronquiolitis Viral/virología , Dermatitis Atópica/inmunología , Eosinofilia/sangre , Adhesión a Directriz , Hospitalización , Humanos , Microbiota/inmunología , Fenotipo , Infecciones por Picornaviridae/tratamiento farmacológico , Infecciones por Picornaviridae/inmunología , Infecciones por Picornaviridae/fisiopatología , Guías de Práctica Clínica como Asunto , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/fisiopatología , Virus Sincitial Respiratorio Humano , Rhinovirus , Células Th2/inmunología , Catelicidinas
10.
Medicina (Kaunas) ; 56(3)2020 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-32120846

RESUMEN

BACKGROUND: Viral infection is the main cause of asthma and COPD (chronic obstructive pulmonary disease) exacerbation and accumulate inflammatory cells to airway tissue. We have reported poly I:C, a mimic product of the virus and ligand of toll-like receptor 3 (TLR3), induced inflammatory chemokines from airway epithelial cells and found prior incubation with corticosteroids diminishes the effect of TLR3 activation. In clinical practice, mild asthma is recommended as-needed budesonide (BUD) when symptoms occur following a viral infection, etc. However, many questions still surround BUD's usefulness if taken after a virus has already infected airway tissue. OBJECTIVE: The aim of this study was to investigate the inhibitory effects of BUD on inflammatory cytokines induced by viral infection. Methods: Normal human bronchial epithelial (NHBE) cells were stimulated with poly I:C or infected with human rhinovirus-16 (HRV16) and BUD was added after the initial stimulation. Expression of both thymic stromal lymphopoietin (TSLP) and CCL26/eotaxin-3 was quantified by real-time RT-PCR and enzyme-linked immunosorbent assay (ELISA), respectively. Knockdown study was performed. Results: Pre-or post-incubation with BUD inhibited both poly I:C- and HRV16-induced mRNAs and proteins of both thymic stromal lymphopoietin (TSLP) and CCL26 with significance. Knockdown of the glucocorticoid receptor diminished these effects of BUD. Under the same conditions of BUD's experiment, post-incubation with neither fluticasone propionate nor dexamethasone suppressed expression of both TSLP and CCL26, which induced by poly I:C. CONCLUSION: Post-addition of BUD inhibited the virus-induced TSLP and CCL26 from the airway epithelial cells. These results suggest that inhalation of BUD after viral infection has beneficial effects on asthma. CONCLUSION: Late addition of BUD may benefit among patient with viral infection and type 2 allergic airway disease such as asthma.


Asunto(s)
Broncodilatadores/farmacología , Budesonida/farmacología , Citocinas/efectos de los fármacos , Infecciones por Picornaviridae/tratamiento farmacológico , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Rhinovirus , Técnicas de Cultivo de Célula , Quimiocina CCL26/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/virología , Humanos , Infecciones por Picornaviridae/virología , Mucosa Respiratoria/citología , Mucosa Respiratoria/virología , Infecciones del Sistema Respiratorio/virología
11.
Am J Respir Cell Mol Biol ; 60(1): 58-67, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30156431

RESUMEN

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.


Asunto(s)
Aldehído Oxidorreductasas/antagonistas & inhibidores , Bronquios/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Inflamación/tratamiento farmacológico , Infecciones por Picornaviridae/tratamiento farmacológico , Rhinovirus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Benzamidas/farmacología , Bronquios/metabolismo , Bronquios/virología , Células Cultivadas , Células Epiteliales/metabolismo , Células Epiteliales/virología , Humanos , Inflamación/metabolismo , Inflamación/virología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Infecciones por Picornaviridae/metabolismo , Infecciones por Picornaviridae/virología , Pirroles/farmacología
12.
Ann Allergy Asthma Immunol ; 123(4): 352-358, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31276807

RESUMEN

OBJECTIVE: To synthesize available data related to the complex associations among viral infections, atopy, and asthma. DATA SOURCES: Key historical articles, articles highlighted in our recent review of most significant recent asthma advancements, and findings from several birth cohorts related to asthma and viral infections were reviewed. In addition, PubMed was searched for review articles and original research related to the associations between viral infection and asthma, using the search words asthma, viral infections, atopy, development of asthma, rhinovirus (RV), and respiratory syncytial virus (RSV). STUDY SELECTIONS: Articles were selected based on novelty and relevance to our topic of interest, the role of asthma and viral infections, and possible mechanisms to explain the association. RESULTS: There is a large body of evidence demonstrating a link between early viral infections (especially RV and RSV) and asthma inception and exacerbations. RV-induced wheezing is an important risk factor for asthma only when atopy is present, with much evidence supporting the idea that sensitization is a risk factor for early RV-induced wheezing, which in turn is a risk factor for asthma. RSV, on the other hand, is a more important risk factor for nonatopic asthma, with severe infections conferring greater risk. CONCLUSION: There are important differences in the development of atopic and nonatopic asthma, with several proposed mechanisms explaining the association between viral infections and the development of asthma and asthma exacerbations. Understanding these complex associations is important for developing asthma prevention strategies and targeted asthma therapies.


Asunto(s)
Asma/inmunología , Infecciones por Picornaviridae/inmunología , Infecciones por Picornaviridae/patología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/patología , Brote de los Síntomas , Asma/patología , Niño , Preescolar , Células Dendríticas/inmunología , Humanos , Neutrófilos/inmunología , Infecciones por Picornaviridae/tratamiento farmacológico , Ruidos Respiratorios/fisiopatología , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Virus Sincitiales Respiratorios/inmunología , Rhinovirus/inmunología
13.
Respirology ; 24(12): 1212-1219, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30989728

RESUMEN

BACKGROUND AND OBJECTIVE: Human rhinovirus (RV) is a common upper and lower respiratory pathogen in lung allograft recipients causing respiratory tract exacerbation and contributing towards allograft dysfunction and long-term lung decline. In this study, we tested the hypothesis that RV could infect both the small and large airways, resulting in significant inflammation. METHODS: Matched large and small airway epithelial cells (AEC) were obtained from five lung allograft recipients. Primary cultures were established, and monolayers were infected with RV1b over time with varying viral titre. Cell viability, receptor expression, viral copy number, apoptotic induction and inflammatory cytokine production were also assessed at each region. Finally, the effect of azithromycin on viral replication, induction of apoptosis and inflammation was investigated. RESULTS: RV infection caused significant cytotoxicity in both large AEC (LAEC) and small AEC (SAEC), and induced a similar apoptotic response in both regions. There was a significant increase in receptor expression in the LAEC only post viral infection. Viral replication was elevated in both LAEC and SAEC, but was not significantly different. Prophylactic treatment of azithromycin reduced viral replication and dampened the production of inflammatory cytokines post-infection. CONCLUSION: Our data illustrate that RV infection is capable of infecting upper and lower AEC, driving cell death and inflammation. Prophylactic treatment with azithromycin was found to mitigate some of the detrimental responses. Findings provide further support for the prophylactic prescription of azithromycin to minimize the impact of RV infection.


Asunto(s)
Células Epiteliales Alveolares , Azitromicina/farmacología , Infecciones por Picornaviridae , Infecciones del Sistema Respiratorio , Rhinovirus , Antibacterianos/farmacología , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citocinas/análisis , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Trasplante de Pulmón/efectos adversos , Infecciones por Picornaviridae/tratamiento farmacológico , Infecciones por Picornaviridae/inmunología , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/virología , Rhinovirus/patogenicidad , Rhinovirus/fisiología , Replicación Viral/efectos de los fármacos
14.
Am J Respir Crit Care Med ; 197(10): 1265-1274, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29466680

RESUMEN

RATIONALE: Immunophenotypes of antiviral responses, and their relationship with asthma, allergy, and lower respiratory tract infections, are poorly understood. OBJECTIVES: We characterized multiple cytokine responses of peripheral blood mononuclear cells to rhinovirus stimulation, and their relationship with clinical outcomes. METHODS: In a population-based birth cohort, we measured 28 cytokines after stimulation with rhinovirus-16 in 307 children aged 11 years. We used machine learning to identify patterns of cytokine responses, and related these patterns to clinical outcomes, using longitudinal models. We also ascertained phytohemagglutinin-induced T-helper cell type 2 (Th2)-cytokine responses (PHA-Th2). MEASUREMENTS AND MAIN RESULTS: We identified six clusters of children based on their rhinovirus-16 responses, which were differentiated by the expression of four cytokine/chemokine groups: interferon-related (IFN), proinflammatory (Inflam), Th2-chemokine (Th2-chem), and regulatory (Reg). Clusters differed in their clinical characteristics. Children with an IFNmodInflamhighestTh2-chemhighestReghighest rhinovirus-16-induced pattern had a PHA-Th2low response, and a very low asthma risk (odds ratio [OR], 0.08; 95% confidence interval [CI], 0.01-0.81; P = 0.03). Two clusters had a high risk of asthma and allergic sensitization, but with different trajectories from infancy to adolescence. The IFNlowestInflamhighTh2-chemlowRegmod cluster exhibited a PHA-Th2lowest response and was associated with early-onset asthma and sensitization, and the highest risk of asthma exacerbations (OR, 1.37; 95% CI, 1.07-1.76; P = 0.014) and lower respiratory tract infection hospitalizations (OR, 2.40; 95% CI, 1.26-4.58; P = 0.008) throughout childhood. In contrast, the IFNhighestInflammodTh2-chemmodReghigh cluster with a rhinovirus-16-cytokine pattern was characterized by a PHA-Th2highest response, and a low prevalence of asthma/sensitization in infancy that increased sharply to become the highest among all clusters by adolescence (but with a low risk of asthma exacerbations). CONCLUSIONS: Early-onset troublesome asthma with early-life sensitization, later-onset milder allergic asthma, and disease protection are each associated with different patterns of rhinovirus-induced immune responses.


Asunto(s)
Antivirales/uso terapéutico , Asma/tratamiento farmacológico , Citocinas/inmunología , Infecciones por Picornaviridae/tratamiento farmacológico , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Rhinovirus/efectos de los fármacos , Rhinovirus/inmunología , Adolescente , Antivirales/inmunología , Niño , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Lactante , Masculino , Infecciones por Picornaviridae/inmunología , Infecciones del Sistema Respiratorio/inmunología
15.
J Allergy Clin Immunol ; 141(4): 1220-1230, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28734844

RESUMEN

BACKGROUND: Human rhinoviruses (HRVs) commonly precipitate asthma exacerbations. Toll-like receptor 3, an innate pattern recognition receptor, is triggered by HRV, driving inflammation that can worsen asthma. OBJECTIVE: We sought to evaluate an inhibitory mAb to Toll-like receptor 3, CNTO3157, on experimental HRV-16 inoculation in healthy subjects and asthmatic patients. METHODS: In this double-blind, multicenter, randomized, parallel-group study in North America and Europe, healthy subjects and patients with mild-to-moderate stable asthma received single or multiple doses of CNTO3157 or placebo, respectively, and were then inoculated with HRV-16 within 72 hours. All subjects were monitored for respiratory symptoms, lung function, and nasal viral load. The primary end point was maximal decrease in FEV1 during 10 days after inoculation. RESULTS: In asthmatic patients (n = 63) CNTO3157 provided no protection against FEV1 decrease (least squares mean: CNTO3157 [n = 30] = -7.08% [SE, 8.15%]; placebo [n = 25] = -5.98% [SE, 8.56%]) or symptoms after inoculation. In healthy subjects (n = 12) CNTO3157 versus placebo significantly attenuated upper (P = .03) and lower (P = .02) airway symptom scores, with area-under-the-curve increases of 9.1 (15.1) versus 34.9 (17.6) and 13.0 (18.4) versus 50.4 (25.9) for the CNTO3157 (n = 8) and placebo (n = 4) groups, respectively, after inoculation. All of the severe and 4 of the nonserious asthma exacerbations occurred while receiving CNTO3157. CONCLUSION: In summary, CNTO3157 was ineffective in attenuating the effect of HRV-16 challenge on lung function, asthma control, and symptoms in asthmatic patients but suppressed cold symptoms in healthy subjects. Other approaches, including blockade of multiple pathways or antiviral agents, need to be sought for this high unmet medical need.


Asunto(s)
Antiinflamatorios/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Asma/tratamiento farmacológico , Asma/virología , Infecciones por Picornaviridae/complicaciones , Rhinovirus , Receptor Toll-Like 3/antagonistas & inhibidores , Adolescente , Adulto , Anciano , Asma/diagnóstico , Asma/inmunología , Progresión de la Enfermedad , Método Doble Ciego , Femenino , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Infecciones por Picornaviridae/tratamiento farmacológico , Infecciones por Picornaviridae/inmunología , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Adulto Joven
16.
Int J Mol Sci ; 20(9)2019 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-31067687

RESUMEN

Rhinovirus (RV) is the predominant virus causing respiratory tract infections. Bronchobini® is a low dose multi component, multi target preparation used to treat inflammatory respiratory diseases such as the common cold, described to ease severity of symptoms such as cough and viscous mucus production. The aim of the study was to assess the efficacy of Bronchobini® in RV infection and to elucidate its mode of action. Therefore, Bronchobini®'s ingredients (BRO) were assessed in an ex vivo model of RV infection using mouse precision-cut lung slices, an organotypic tissue capable to reflect the host immune response to RV infection. Cytokine profiles were assessed using enzyme-linked immunosorbent assay (ELISA) and mesoscale discovery (MSD). Gene expression analysis was performed using Affymetrix microarrays and ingenuity pathway analysis. BRO treatment resulted in the significant suppression of RV-induced antiviral and pro-inflammatory cytokine release. Transcriptome analysis revealed a multifactorial mode of action of BRO, with a strong inhibition of the RV-induced pro-inflammatory and antiviral host response mediated by nuclear factor kappa B (NFkB) and interferon signaling pathways. Interestingly, this was due to priming of these pathways in the absence of virus. Overall, BRO exerted its beneficial anti-inflammatory effect by priming the antiviral host response resulting in a reduced inflammatory response to RV infection, thereby balancing an otherwise excessive inflammatory response.


Asunto(s)
Antivirales/farmacología , Inductores de Interferón/farmacología , Interferones/metabolismo , Pulmón/efectos de los fármacos , Infecciones por Picornaviridae/tratamiento farmacológico , Extractos Vegetales/farmacología , Transcriptoma , Animales , Antivirales/uso terapéutico , Femenino , Inductores de Interferón/uso terapéutico , Pulmón/metabolismo , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Infecciones por Picornaviridae/inmunología , Infecciones por Picornaviridae/virología , Extractos Vegetales/uso terapéutico , Rhinovirus/efectos de los fármacos , Rhinovirus/patogenicidad , Transducción de Señal
17.
Am J Respir Cell Mol Biol ; 59(6): 713-722, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30084659

RESUMEN

Human rhinovirus (RV) infections are a significant risk factor for exacerbations of asthma and chronic obstructive pulmonary disease. Thus, approaches to prevent RV infection in such patients would give significant benefit. Through RNA interference library screening, we identified lanosterol synthase (LSS), a component of the cholesterol biosynthetic pathway, as a novel regulator of RV replication in primary normal human bronchial epithelial cells. Selective knock down of LSS mRNA with short interfering RNA inhibited RV2 replication in normal human bronchial epithelial cells. Small molecule inhibitors of LSS mimicked the effect of LSS mRNA knockdown in a concentration-dependent manner. We further demonstrated that the antiviral effect is not dependent on a reduction in total cellular cholesterol but requires a 24-hour preincubation with the LSS inhibitor. The rank order of antiviral potency of the LSS inhibitors used was consistent with LSS inhibition potency; however, all compounds showed remarkably higher potency against RV compared with the LSS enzyme potency. We showed that LSS inhibition led to an induction of 24(S),25 epoxycholesterol, an important regulator of the sterol pathway. We also demonstrated that LSS inhibition led to a profound increase in expression of the innate antiviral defense protein, IFN-ß. We found LSS to be a novel regulator of RV replication and innate antiviral immunity and identified a potential molecular mechanism for this effect, via induction of 24(S),25 epoxycholesterol. Inhibition of LSS could therefore be a novel therapeutic target for prevention of RV-induced exacerbations.


Asunto(s)
Antivirales/farmacología , Bronquios/inmunología , Células Epiteliales/inmunología , Inmunidad Innata/inmunología , Transferasas Intramoleculares/metabolismo , Infecciones por Picornaviridae/inmunología , Rhinovirus/inmunología , Replicación Viral/inmunología , Bronquios/efectos de los fármacos , Bronquios/virología , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/virología , Humanos , Inmunidad Innata/efectos de los fármacos , Transferasas Intramoleculares/antagonistas & inhibidores , Transferasas Intramoleculares/genética , Infecciones por Picornaviridae/tratamiento farmacológico , Infecciones por Picornaviridae/virología , ARN Interferente Pequeño/genética , Rhinovirus/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Replicación Viral/efectos de los fármacos
18.
J Infect Chemother ; 24(9): 734-738, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29895452

RESUMEN

Although viruses are the major pathogen that causes upper respiratory tract infection (URTI) and acute bronchitis, antibiotics have been prescribed. This was a prospective observational study in influenza epidemics that enrolled adult outpatients who visited a hospital with respiratory tract infection symptoms. In this study, we evaluated the usefulness of FilmArray respiratory panel (RP). Fifty patients were enrolled. FilmArray RP detected the pathogens in 28 patients. The common pathogens were influenza virus (n = 14), respiratory syncytial virus (n = 6), and human rhinovirus (n = 6). Of the 14 patients with influenza virus, 6 were negative for the antigen test. The physicians diagnosed and treated the patients without the result of FilmArray in this study. Of the patients with positive FilmArray RP, 9 were treated with antibiotics; however, bacteria were detected in only 3 patients. By implementing FilmArray RP, URTI and acute bronchitis would be precisely diagnosed, and inappropriate use of antibiotics can be reduced.


Asunto(s)
Reacción en Cadena de la Polimerasa Multiplex/métodos , Infecciones del Sistema Respiratorio/diagnóstico , Infecciones del Sistema Respiratorio/virología , Enfermedad Aguda/terapia , Antibacterianos/efectos adversos , Antibacterianos/uso terapéutico , Femenino , Humanos , Gripe Humana/diagnóstico , Gripe Humana/tratamiento farmacológico , Gripe Humana/virología , Masculino , Persona de Mediana Edad , Pacientes Ambulatorios , Infecciones por Picornaviridae/diagnóstico , Infecciones por Picornaviridae/tratamiento farmacológico , Infecciones por Picornaviridae/virología , Estudios Prospectivos , Infecciones por Virus Sincitial Respiratorio/diagnóstico , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/efectos de los fármacos , Virus Sincitiales Respiratorios/genética , Infecciones del Sistema Respiratorio/tratamiento farmacológico
19.
J Infect Dis ; 215(7): 1102-1106, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28368456

RESUMEN

BACKGROUND: Respiratory syncytial virus (RSV) and human rhinovirus (HRV) are the most common viruses associated with acute respiratory tract infections in infancy. Viral interference is important in understanding respiratory viral circulation and the impact of vaccines. METHODS: To study viral interference, we evaluated cases of RSV and HRV codetection by polymerase chain reaction in 2 prospective birth cohort studies (the Infant Susceptibility to Pulmonary Infections and Asthma Following RSV Exposure [INSPIRE] study and the Tennessee Children's Respiratory Initiative [TCRI]) and a double-blinded, randomized, controlled trial (MAKI), using adjusted multivariable regression analyses. RESULTS: Among 3263 respiratory tract samples, 24.5% (798) and 37.3% (1216) were RSV and HRV positive, respectively. The odds of HRV infection were significantly lower in RSV-infected infants in all cohorts, with adjusted odds ratios of 0.30 (95% confidence interval [CI], .22-.40 in the INSPIRE study, 0.18 (95% CI, .11-.28) in the TCRI (adjusted for disease severity), and 0.34 (95% CI, .16-.72) in the MAKI trial. HRV infection was significantly more common among infants administered RSV immunoprophylaxis, compared with infants who did not receive immunoprophylaxis (OR, 1.65; 95% CI, 1.65-2.39). CONCLUSIONS: A negative association of RSV on HRV codetection was consistently observed across populations, seasons, disease severity, and geographical regions. Suppressing RSV infection by RSV immunoprophylaxis might increase the risk of having HRV infection.


Asunto(s)
Coinfección/epidemiología , Coinfección/virología , Infecciones por Picornaviridae/epidemiología , Infecciones por Virus Sincitial Respiratorio/epidemiología , Antivirales/uso terapéutico , Susceptibilidad a Enfermedades , Método Doble Ciego , Femenino , Humanos , Lactante , Tiempo de Internación , Modelos Logísticos , Masculino , Análisis Multivariante , Palivizumab/uso terapéutico , Infecciones por Picornaviridae/tratamiento farmacológico , Reacción en Cadena de la Polimerasa , Estudios Prospectivos , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Virus Sincitial Respiratorio Humano , Rhinovirus , Estados Unidos
20.
J Virol ; 90(21): 9725-9732, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27535044

RESUMEN

Equine rhinitis A virus (ERAV) is a picornavirus associated with respiratory disease in horses and is genetically closely related to foot-and-mouth disease virus (FMDV), the prototype aphthovirus. ERAV has recently gained interest as an FMDV alternative for the study of aphthovirus biology, including cell entry and uncoating or antiviral testing. As described for FMDV, current data support that acidic pH inside cellular endosomes triggers ERAV uncoating. In order to provide further insights into aphthovirus uncoating mechanism, we have isolated a panel of ERAV mutants with altered acid sensitivity and that differed on their degree of sensitivity to the inhibition of endosome acidification. These results provide functional evidence of the involvement of acidic pH on ERAV uncoating within endosomes. Remarkably, all amino acid substitutions found in acid-labile or acid-resistant ERAVs were located in the capsid protein VP3, indicating that this protein plays a pivotal role for the control of pH stability of the ERAV capsid. Moreover, all amino acid substitutions mapped at the intraprotomer interface between VP3 and VP2 or between VP3 and the N terminus of VP1. These results expand our knowledge on the regions that regulate the acid stability of aphthovirus capsid and should be taken into account when using ERAV as a surrogate of FMDV. IMPORTANCE: The viral capsid constitutes a sort of dynamic nanomachine that protects the viral genome against environmental assaults while accomplishing important functions such as receptor attachment for viral entry or genome release. We have explored the molecular determinants of aphthovirus capsid stability by isolating and characterizing a panel of equine rhinitis A virus mutants that differed on their acid sensitivity. All the mutations were located within a specific region of the capsid, the intraprotomer interface among capsid proteins, thus providing new insights into the regions that control the acid stability of aphthovirus capsid. These findings could positively contribute to the development of antiviral approaches targeting aphthovirus uncoating or the refinement of vaccine strategies based on capsid stabilization.


Asunto(s)
Ácidos/metabolismo , Aphthovirus/genética , Proteínas de la Cápside/genética , Caballos/virología , Sustitución de Aminoácidos/genética , Animales , Antivirales/farmacología , Aphthovirus/efectos de los fármacos , Cápside/efectos de los fármacos , Endosomas/virología , Virus de la Fiebre Aftosa/efectos de los fármacos , Genoma Viral/genética , Concentración de Iones de Hidrógeno , Mutación/genética , Infecciones por Picornaviridae/tratamiento farmacológico , Infecciones por Picornaviridae/virología , Internalización del Virus/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda