Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Mol Immunol ; 138: 20-30, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34332182

RESUMEN

Respiratory syncytial virus (RSV) infection in early life is associated strongly with the subsequent development and exacerbation of asthma, however, the mechanism is still ambiguous. In this study, we identified that RSV nonstructural protein (NS) 1 plays a critical role. Plasmid-mediated overexpression of NS1 induced significant airway hyperresponsiveness, eosinophilia, and mucus hyperproduction in mice. In the pNS1 group, there were markedly elevated proportions of Th2 and Th17 cells, while Th1 and Foxp3+ regulatory T cells (Tregs) significantly declined compared with the control group. Serum concentrations of interleukin (IL)-4, IL-5, IL-6, IL-17, transforming growth factor-beta, and tumor necrosis factor-alpha increased but levels of interferon-gamma and interleukin-10 declined in pNS1 group. Besides, NS1 caused a significant rise of serum thymic stromal lymphopoietin (TSLP) and OX40L levels, and a neutralizing mAb anti-OX40L was capable of promoting RSV clearance and attenuating the airway allergic inflammation caused by pNS1. Otherwise, OX40L-blocking counteracts the inhibitory effect of pNS1 on Tregs in the spleen. RSV NS1 caused elevated levels of phospho-AKT, phospho-mTOR, and phospho-S6K1, which were partially attenuated by anti-OX40L. Moreover, a specific inhibitor of mTORC1 significantly relieved the inhibition of Foxp3 expression and Tregs differentiation. Together, the data indicate that RSV NS1 protein breaks immune tolerance and induces airway inflammation and hyperresponsiveness in mice. In this process, NS1-stimulated TSLP and OX40L play a major role by inhibiting the induction of Tregs, which is at least partially mediated by modulating AKT-mTOR signaling pathways.


Asunto(s)
Tolerancia Inmunológica/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Proteínas no Estructurales Virales/inmunología , Animales , Citocinas/inmunología , Regulación hacia Abajo , Femenino , Ratones , Ratones Endogámicos BALB C , Ligando OX40/inmunología , Receptores OX40/inmunología , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/virología , Infecciones por Virus Sincitial Respiratorio/complicaciones , Virus Sincitial Respiratorio Humano/inmunología , Serina-Treonina Quinasas TOR/inmunología , Linfopoyetina del Estroma Tímico
2.
Viruses ; 13(5)2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33922096

RESUMEN

Respiratory virus infections can have long-term effects on lung function that persist even after the acute responses have resolved. Numerous studies have linked severe early childhood infection with respiratory syncytial virus (RSV) to the development of wheezing and asthma, although the underlying mechanisms connecting these observations remain unclear. Here, we examine airway hyperresponsiveness (AHR) that develops in wild-type mice after recovery from symptomatic but sublethal infection with the natural rodent pathogen, pneumonia virus of mice (PVM). We found that BALB/c mice respond to a limited inoculum of PVM with significant but reversible weight loss accompanied by virus replication, acute inflammation, and neutrophil recruitment to the airways. At day 21 post-inoculation, virus was no longer detected in the airways and the acute inflammatory response had largely resolved. However, and in contrast to most earlier studies using the PVM infection model, all mice survived the initial infection and all went on to develop serum anti-PVM IgG antibodies. Furthermore, using both invasive plethysmography and precision-cut lung slices, we found that these mice exhibited significant airway hyperresponsiveness at day 21 post-inoculation that persisted through day 45. Taken together, our findings extend an important and versatile respiratory virus infection model that can now be used to explore the role of virions and virion clearance as well as virus-induced inflammatory mediators and their signaling pathways in the development and persistence of post-viral AHR and lung dysfunction.


Asunto(s)
Virus de la Neumonía Murina/inmunología , Infecciones por Pneumovirus/complicaciones , Infecciones por Pneumovirus/veterinaria , Hipersensibilidad Respiratoria/etiología , Animales , Anticuerpos Antivirales/inmunología , Humanos , Pulmón/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Virus de la Neumonía Murina/fisiología , Infecciones por Pneumovirus/inmunología , Infecciones por Pneumovirus/virología , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/virología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/inmunología , Virus Sincitial Respiratorio Humano/fisiología
3.
Am J Respir Cell Mol Biol ; 62(3): 310-318, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31533004

RESUMEN

Rhinovirus (RV) exposure evokes exacerbations of asthma that markedly impact morbidity and mortality worldwide. The mechanisms by which RV induces airway hyperresponsiveness (AHR) or by which specific RV serotypes differentially evoke AHR remain unknown. We posit that RV infection evokes AHR and inflammatory mediator release, which correlate with degrees of RV infection. Furthermore, we posit that rhinovirus C-induced AHR requires paracrine or autocrine mediator release from epithelium that modulates agonist-induced calcium mobilization in human airway smooth muscle. In these studies, we used an ex vivo model to measure bronchoconstriction and mediator release from infected airways in human precision cut lung slices to understand how RV exposure alters airway constriction. We found that rhinovirus C15 (RV-C15) infection augmented carbachol-induced airway narrowing and significantly increased release of IP-10 (IFN-γ-induced protein 10) and MIP-1ß (macrophage inflammatory protein-1ß) but not IL-6. RV-C15 infection of human airway epithelial cells augmented agonist-induced intracellular calcium flux and phosphorylation of myosin light chain in co-cultured human airway smooth muscle to carbachol, but not after histamine stimulation. Our data suggest that RV-C15-induced structural cell inflammatory responses are associated with viral load but that inflammatory responses and alterations in agonist-mediated constriction of human small airways are uncoupled from viral load of the tissue.


Asunto(s)
Señalización del Calcio , Infecciones por Enterovirus/fisiopatología , Enterovirus/fisiología , Músculo Liso/virología , Hipersensibilidad Respiratoria/etiología , Asma/virología , Carbacol/farmacología , Células Cultivadas , Quimiocina CXCL10/metabolismo , Enterovirus/genética , Enterovirus/aislamiento & purificación , Infecciones por Enterovirus/virología , Histamina/farmacología , Humanos , Mediadores de Inflamación/metabolismo , Contracción Muscular/efectos de los fármacos , Músculo Liso/fisiopatología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , ARN Viral/análisis , Hipersensibilidad Respiratoria/virología , Carga Viral
4.
Viruses ; 11(6)2019 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-31195744

RESUMEN

Rhinovirus (RV) is an RNA virus that causes more than 50% of upper respiratory tract infections in humans worldwide. Together with Respiratory Syncytial Virus, RV is one of the leading causes of viral bronchiolitis in infants and the most common virus associated with wheezing in children aged between one and two years. Because of its tremendous genetic diversity (>150 serotypes), the recurrence of RV infections each year is quite typical. Furthermore, because of its broad clinical spectrum, the clinical variability as well as the pathogenesis of RV infection are nowadays the subjects of an in-depth examination and have been the subject of several studies in the literature. In fact, the virus is responsible for direct cell cytotoxicity in only a small way, and it is now clearer than ever that it may act indirectly by triggering the release of active mediators by structural and inflammatory airway cells, causing the onset and/or the acute exacerbation of asthmatic events in predisposed children. In the present review, we aim to summarize the RV infection's epidemiology, pathogenetic hypotheses, and available treatment options as well as its correlation with respiratory morbidity and mortality in the pediatric population.


Asunto(s)
Infecciones por Picornaviridae , Hipersensibilidad Respiratoria/virología , Rhinovirus , Inmunidad Adaptativa , Antivirales/uso terapéutico , Asma/etiología , Asma/virología , Bronquiolitis/microbiología , Bronquiolitis/virología , Niño , Salud Infantil , Humanos , Inmunidad Celular , Lactante , Inflamación/inmunología , Inflamación/virología , Infecciones por Picornaviridae/complicaciones , Infecciones por Picornaviridae/epidemiología , Infecciones por Picornaviridae/terapia , Infecciones por Picornaviridae/transmisión , Hipersensibilidad Respiratoria/inmunología , Ruidos Respiratorios/etiología , Rhinovirus/clasificación , Rhinovirus/efectos de los fármacos , Rhinovirus/inmunología , Rhinovirus/patogenicidad , Serogrupo , Vacunas Virales
5.
Pediatr Res ; 86(1): 39-46, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30986815

RESUMEN

BACKGROUND: Environmental tobacco smoke (ETS) is a known risk factor for severe respiratory syncytial virus (RSV) infections, yet the mechanisms of ETS/RSV comorbidity are largely unknown. Cystathionine γ-lyase regulates important physiological functions of the respiratory tract. METHODS: We used mice genetically deficient in the cystathionine γ-lyase enzyme (CSE), the major H2S-generating enzyme in the lung to determine the contribution of H2S to airway disease in response to side-stream tobacco smoke (TS), and to TS/RSV co-exposure. RESULTS: Following a 2-week period of exposure to TS, CSE-deficient mice (KO) showed a dramatic increase in airway hyperresponsiveness (AHR) to methacholine challenge, and greater airway cellular inflammation, compared with wild-type (WT) mice. TS-exposed CSE KO mice that were subsequently infected with RSV exhibited a more severe clinical disease, airway obstruction and AHR, enhanced viral replication, and lung inflammation, compared with TS-exposed RSV-infected WT mice. TS-exposed RSV-infected CSE KO mice had also a significant increase in the number of neutrophils in bronchoalveolar lavage fluid and increased levels of inflammatory cytokines and chemokines. CONCLUSION: This study demonstrates the critical contribution of the H2S-generating pathway to airway reactivity and disease following exposure to ETS alone or in combination with RSV infection.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/fisiopatología , Cistationina gamma-Liasa/deficiencia , Pulmón/fisiopatología , Pulmón/virología , Hipersensibilidad Respiratoria/complicaciones , Infecciones por Virus Sincitial Respiratorio/complicaciones , Contaminación por Humo de Tabaco/efectos adversos , Animales , Líquido del Lavado Bronquioalveolar , Citocinas/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Sulfuro de Hidrógeno/química , Inflamación/etiología , Masculino , Cloruro de Metacolina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/citología , Hipersensibilidad Respiratoria/virología , Virus Sincitiales Respiratorios
6.
Respir Res ; 19(1): 208, 2018 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-30373568

RESUMEN

Rhinovirus (RV) exposure has been implicated in childhood development of wheeze evoking asthma and exacerbations of underlying airways disease. Studies such as the Copenhagen Prospective Studies on Asthma in Childhood (COPSAC) and Childhood Origins of ASThma (COAST) have identified RV as a pathogen inducing severe respiratory disease. RVs also modulate airway hyperresponsiveness (AHR), a key characteristic of such diseases. Although potential factors underlying mechanisms by which RV induces AHR have been postulated, the precise mechanisms of AHR following RV exposure remain elusive.A challenge to RV-related research stems from inadequate models for study. While human models raise ethical concerns and are relatively difficult in terms of subject recruitment, murine models are limited by susceptibility of infection to the relatively uncommon minor group (RV-B) serotypes, strains that are generally associated with infrequent clinical respiratory virus infections. Although a transgenic mouse strain that has been developed has enhanced susceptibility for infection with the common major group (RV-A) serotypes, few studies have focused on RV in the context of allergic airways disease rather than understanding RV-induced AHR. Recently, the receptor for the virulent RV-C CDHR3, was identified, but a dearth of studies have examined RV-C-induced effects in humans.Currently, the mechanisms by which RV infections modulate airway smooth muscle (ASM) shortening or excitation-contraction coupling remain elusive. Further, only one study has investigated the effects of RV on bronchodilatory mechanisms, with only speculation as to mechanisms underlying RV-mediated modulation of bronchoconstriction.


Asunto(s)
Hipersensibilidad Respiratoria/fisiopatología , Hipersensibilidad Respiratoria/virología , Rhinovirus/aislamiento & purificación , Rhinovirus/fisiología , Contaminantes Atmosféricos/efectos adversos , Animales , Asma/epidemiología , Asma/fisiopatología , Asma/virología , Técnicas de Cocultivo , Humanos , Hipersensibilidad Respiratoria/epidemiología
8.
J Allergy Clin Immunol ; 142(4): 1206-1217.e5, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29269317

RESUMEN

BACKGROUND: Viral respiratory tract infections increase the risk of development and exacerbation of atopic disease. Previously, we demonstrated the requirement for a neutrophil (PMN) subset expressing CD49d to drive development of postviral atopic airway disease in mice. OBJECTIVE: We sought to determine whether human CD49d+ PMNs are present in the nasal mucosa during acute viral respiratory tract infections and further characterize this PMN subset in human subjects and mice. METHODS: Sixty subjects (5-50 years old) were enrolled within 4 days of acute onset of upper respiratory symptoms. Nasal lavage for flow cytometry and nasal swabs for viral PCR were performed at enrollment and during convalescence. The Sendai virus mouse model was used to investigate the phenotype and functional relevance of CD49d+ PMNs. RESULTS: CD49d+ PMN frequency was significantly higher in nasal lavage fluid during acute respiratory symptoms in all subjects (2.9% vs 1.0%, n = 42, P < .001). In mice CD49d+ PMNs represented a "proatopic" neutrophil subset that expressed cysteinyl leukotriene receptor 1 (CysLTR1) and produced TNF, CCL2, and CCL5. Inhibition of CysLTR1 signaling in the first days of a viral respiratory tract infection was sufficient to reduce accumulation of CD49d+ PMNs in the lungs and development of postviral atopic airway disease. Similar to the mouse, human CD49d+ PMNs isolated from nasal lavage fluid during a viral respiratory tract infection expressed CysLTR1. CONCLUSION: CD49d and CysLTR1-coexpressing PMNs are present during symptoms of an acute viral respiratory tract infection in human subjects. Further study is needed to examine selective targeting of proatopic neutrophils as a potential therapeutic strategy to prevent development of postviral atopic airway disease.


Asunto(s)
Integrina alfa4/inmunología , Mucosa Nasal/inmunología , Neutrófilos/inmunología , Receptores de Leucotrienos/inmunología , Hipersensibilidad Respiratoria/inmunología , Infecciones del Sistema Respiratorio/inmunología , Infecciones por Respirovirus/inmunología , Adolescente , Adulto , Animales , Niño , Preescolar , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Mucosa Nasal/citología , Mucosa Nasal/virología , Hipersensibilidad Respiratoria/virología , Infecciones del Sistema Respiratorio/virología , Infecciones por Respirovirus/virología , Virus Sendai , Adulto Joven
9.
Int J Mol Sci ; 18(10)2017 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-29065558

RESUMEN

The original hygiene hypothesis declares "more infections in early childhood protect against later atopy". According to the hygiene hypothesis, the increased incidence of allergic disorders in developed countries is explained by the decrease of infections. Epithelial cells and dendritic cells play key roles in bridging the innate and adaptive immune systems. Among the various pattern-recognition receptor systems of epithelial cells and dendritic cells, including toll-like receptors (TLRs), nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs) and others, TLRs are the key systems of immune response regulation. In humans, TLRs consist of TLR1 to TLR10. They regulate cellular responses through engagement with TLR ligands, e.g., lipopolysaccharides (LPS) acts through TLR4 and dsRNA acts through TLR3, but there are certain common components between these two TLR pathways. dsRNA activates epithelial cells and dendritic cells in different directions, resulting in allergy-related Th2-skewing tendency in epithelial cells, and Th1-skewing tendency in dendritic cells. The Th2-skewing effect by stimulation of dsRNA on epithelial cells could be suppressed by the presence of LPS above some threshold. When LPS level decreases, the Th2-skewing effect increases. It may be via these interrelated networks and related factors that LPS modifies the allergic responses and provides a plausible mechanism of the hygiene hypothesis. Several hygiene hypothesis-related phenomena, seemingly conflicting, are also discussed in this review, along with their proposed mechanisms.


Asunto(s)
Hipótesis de la Higiene , Lipopolisacáridos/inmunología , Infecciones por Virus ARN/complicaciones , Hipersensibilidad Respiratoria/etiología , Animales , Células Dendríticas , Células Epiteliales , Humanos , Infecciones por Virus ARN/inmunología , Virus ARN , ARN Bicatenario , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/prevención & control , Hipersensibilidad Respiratoria/virología
10.
PLoS One ; 12(7): e0181425, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28742120

RESUMEN

Asthma exacerbation can be a life-threatening condition, and is most often triggered by common respiratory viruses. Poor asthma control and worsening of respiratory function is associated with increased airway inflammation, including eosinophilia. Prevention of asthma exacerbation relies on treatment with corticosteroids, which preferentially inhibit allergic inflammation like eosinophils. Human studies demonstrate that inactivated virus can trigger eosinophil activation in vitro through antigen presentation and memory CD4+ lymphocytes. We hypothesized that animals with immunologic memory to a respiratory virus would also develop airway hyperresponsiveness in response to a UV-inactivated form of the virus if they have pre-existing allergic airway inflammation. Guinea pigs were ovalbumin-sensitized, infected with live parainfluenza virus (PIV), aerosol-challenged with ovalbumin, and then re-inoculated 60 days later with live or UV-inactivated PIV. Some animals were either treated with dexamethasone prior to the second viral exposure. Lymphocytes were isolated from parabronchial lymph nodes to confirm immunologic memory to the virus. Airway reactivity was measured and inflammation was assessed using bronchoalveolar lavage and lung histology. The induction of viral immunologic memory was confirmed in infected animals. Allergen sensitized and challenged animals developed airway hyperreactivity with eosinophilic airway inflammation when re-exposed to UV-inactivated PIV, while non-sensitized animals did not. Airway hyperreactivity in the sensitized animals was inhibited by pre-treatment with dexamethasone. We suggest that the response of allergic inflammation to virus antigen is a significant factor causing asthma exacerbation. We propose that this is one mechanism explaining how corticosteroids prevent virus-induced asthma attack.


Asunto(s)
Asma/virología , Virus de la Parainfluenza 1 Humana/inmunología , Hipersensibilidad Respiratoria/virología , Infecciones por Respirovirus/complicaciones , Animales , Antiinflamatorios/uso terapéutico , Asma/tratamiento farmacológico , Asma/inmunología , Dexametasona/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Cobayas , Humanos , Memoria Inmunológica/efectos de los fármacos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/virología , Linfocitos/inmunología , Linfocitos/virología , Hipersensibilidad Respiratoria/tratamiento farmacológico , Hipersensibilidad Respiratoria/inmunología , Infecciones por Respirovirus/tratamiento farmacológico , Infecciones por Respirovirus/inmunología
11.
J Immunol ; 199(4): 1308-1318, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28701507

RESUMEN

Early-life respiratory viral infection is a risk factor for asthma development. Rhinovirus (RV) infection of 6-d-old mice, but not mature mice, causes mucous metaplasia and airway hyperresponsiveness that are associated with the expansion of lung type 2 innate lymphoid cells (ILC2s) and are dependent on IL-13 and the innate cytokine IL-25. However, contributions of the other innate cytokines, IL-33 and thymic stromal lymphopoietin (TSLP), to the observed asthma-like phenotype have not been examined. We reasoned that IL-33 and TSLP expression are also induced by RV infection in immature mice and are required for maximum ILC2 expansion and mucous metaplasia. We inoculated 6-d-old BALB/c (wild-type) and TSLP receptor-knockout mice with sham HeLa cell lysate or RV. Selected mice were treated with neutralizing Abs to IL-33 or recombinant IL-33, IL-25, or TSLP. ILC2s were isolated from RV-infected immature mice and treated with innate cytokines ex vivo. RV infection of 6-d-old mice increased IL-33 and TSLP protein abundance. TSLP expression was localized to the airway epithelium, whereas IL-33 was expressed in epithelial and subepithelial cells. RV-induced mucous metaplasia, ILC2 expansion, airway hyperresponsiveness, and epithelial cell IL-25 expression were attenuated by anti-IL-33 treatment and in TSLP receptor-knockout mice. Administration of intranasal IL-33 and TSLP was sufficient for mucous metaplasia. Finally, TSLP was required for maximal ILC2 gene expression in response to IL-25 and IL-33. The generation of mucous metaplasia in immature RV-infected mice involves a complex interplay among the innate cytokines IL-25, IL-33, and TSLP.


Asunto(s)
Citocinas/inmunología , Interleucina-33/inmunología , Interleucinas/inmunología , Activación de Linfocitos , Linfocitos/fisiología , Metaplasia/inmunología , Infecciones por Picornaviridae/inmunología , Rhinovirus/inmunología , Factores de Edad , Animales , Asma/inmunología , Asma/virología , Citocinas/genética , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/virología , Inmunoglobulinas/genética , Inmunoglobulinas/inmunología , Inmunoglobulinas/metabolismo , Interleucina-33/genética , Interleucinas/genética , Linfocitos/inmunología , Metaplasia/patología , Metaplasia/virología , Ratones , Ratones Noqueados , Membrana Mucosa/inmunología , Membrana Mucosa/patología , Infecciones por Picornaviridae/virología , Receptores de Citocinas/genética , Receptores de Citocinas/inmunología , Receptores de Citocinas/metabolismo , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/metabolismo , Hipersensibilidad Respiratoria/virología , Linfopoyetina del Estroma Tímico
12.
J Leukoc Biol ; 101(2): 519-529, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27566834

RESUMEN

Respiratory syncytial virus (RSV)-related hospitalization during infancy is strongly associated with the subsequent development of asthma. Early life RSV infection results in a Th2-biased immune response, which is also typical of asthma. Murine models of neonatal RSV infection have been developed to examine the possible contribution of RSV-driven Th2 responses to the development of airway hyper-responsiveness later in childhood. We have investigated the ability of a cell-penetrating STAT6 inhibitory peptide (STAT6-IP), when delivered selectively during neonatal RSV infection, to modify pathogenesis induced upon secondary RSV reinfection of adults 6 wk later. Neonatal STAT6-IP treatment inhibited the development of airway hyper-responsiveness (AHR) and significantly reduced lung eosinophilia and collagen deposition in adult mice following RSV reinfection. STAT6-IP-treated, RSV-infected neonates had reduced levels of both IL-4 and alternatively activated macrophages (AAMs) in the lungs. Our findings suggest that targeting STAT6 activity at the time of early-life RSV infection may effectively reduce the risk of subsequent asthma development.


Asunto(s)
Pulmón/patología , Pulmón/virología , Péptidos/farmacología , Infecciones por Virus Sincitial Respiratorio/complicaciones , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/efectos de los fármacos , Factor de Transcripción STAT6/antagonistas & inhibidores , Envejecimiento/patología , Animales , Animales Recién Nacidos , Recuento de Células , Colágeno/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Interleucina-17/metabolismo , Interleucina-33/metabolismo , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/patología , Activación de Macrófagos/efectos de los fármacos , Masculino , Ratones Endogámicos BALB C , Hipersensibilidad Respiratoria/complicaciones , Hipersensibilidad Respiratoria/patología , Hipersensibilidad Respiratoria/virología , Infecciones por Virus Sincitial Respiratorio/patología , Factor de Transcripción STAT6/metabolismo , Factores de Tiempo , Linfopoyetina del Estroma Tímico
13.
PLoS One ; 11(5): e0155526, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27213574

RESUMEN

The effect of respiratory tract viral infection on evoked cough in guinea pigs was evaluated. Guinea pigs were inoculated intranasally with either parainfluenza type 3 (PIV3) and cough was quantified in conscious animals. The guinea pigs infected with PIV3 (day 4) coughed nearly three times more than those treated with the viral growth medium in response to capsaicin, citric acid, and bradykinin. Since capsaicin, citric acid, and bradykinin evoked coughing in guinea pigs can be inhibited by drugs that antagonize the transient receptor potential cation channel, subfamily V, member 1 (TRPV1), it was reasoned that the virally-induced hypertussive state may involve alterations in TPRV1 activity. PIV3 infection caused a phenotypic switch in tracheal nodose Aδ "cough receptors" such that nearly 50% of neurons began to express, de novo, TRPV1 mRNA. There was also an increase TRPV1 expression in jugular C-fiber neurons as determined by qPCR. It has previously been reported that tracheal-specific nodose neurons express the BDNF receptor TrkB and jugular neurons express the NGF receptor TrkA. Jugular neurons also express the artemin receptor GFRα3. All these neurotrophic factors have been associated with increases in TRPV1 expression. In an ex vivo perfused guinea pig tracheal preparation, we demonstrated that within 8 h of PIV3 infusion there was no change in NGF mRNA expression, but there was nearly a 10-fold increase in BDNF mRNA in the tissue, and a small but significant elevation in the expression of artemin mRNA. In summary, PIV3 infection leads to elevations in TRPV1 expression in the two key cough evoking nerve subtypes in the guinea pig trachea, and this is associated with a hypertussive state with respect to various TRPV1 activating stimuli.


Asunto(s)
Tos/virología , Virus de la Parainfluenza 3 Humana/fisiología , Infecciones por Paramyxoviridae/complicaciones , Hipersensibilidad Respiratoria/virología , Animales , Tos/genética , Tos/inmunología , Expresión Génica , Cobayas , Masculino , Fibras Nerviosas Amielínicas/fisiología , Fibras Nerviosas Amielínicas/virología , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Ganglio Nudoso/inmunología , Ganglio Nudoso/virología , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/inmunología , Infecciones por Paramyxoviridae/virología , Hipersensibilidad Respiratoria/complicaciones , Hipersensibilidad Respiratoria/genética , Hipersensibilidad Respiratoria/inmunología , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Tráquea/inmunología , Tráquea/inervación , Tráquea/metabolismo , Tráquea/virología
14.
Trends Pharmacol Sci ; 37(4): 252-261, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26833119

RESUMEN

Lung diseases, such as asthma and chronic obstructive pulmonary disease (COPD), are highly prevalent worldwide. One of the major factors that limits the efficacy of current medication in these patients are viral infections, leading to exacerbations of symptoms and decreased quality of life. Current pharmacological strategies targeting virus-induced lung disease are problematic due to antiviral resistance and the requirement for strain-specific vaccination. Thus, new therapeutic strategies are urgently required. In this Opinion article, we provide state-of-the-art evidence from humans and preclinical animal models implicating the interleukin (IL)-33/IL-13 axis in virus-induced lung disease. Thus, targeting the IL-33/IL-13 axis may be a feasible way to overcome the limitations of current therapy used to treat virus-induced exacerbations of lung disease.


Asunto(s)
Antivirales/farmacología , Asma/tratamiento farmacológico , Interleucina-13/antagonistas & inhibidores , Interleucina-33/antagonistas & inhibidores , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Virosis/tratamiento farmacológico , Animales , Asma/inmunología , Asma/virología , Humanos , Inmunidad Innata , Interleucina-13/inmunología , Interleucina-33/inmunología , Terapia Molecular Dirigida , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/virología , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/virología , Transducción de Señal , Virosis/inmunología
15.
PLoS One ; 10(8): e0135970, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26284919

RESUMEN

Increased lung levels of matrix metalloproteinase 9 (MMP9) are frequently observed during respiratory syncytial virus (RSV) infection and elevated MMP9 concentrations are associated with severe disease. However little is known of the functional role of MMP9 during lung infection with RSV. To determine whether MMP9 exerted direct antiviral potential, active MMP9 was incubated with RSV, which showed that MMP9 directly prevented RSV infectivity to airway epithelial cells. Using knockout mice the effect of the loss of Mmp9 expression was examined during RSV infection to demonstrate MMP9's role in viral clearance and disease progression. Seven days following RSV infection, Mmp9-/- mice displayed substantial weight loss, increased RSV-induced airway hyperresponsiveness (AHR) and reduced clearance of RSV from the lungs compared to wild type mice. Although total bronchoalveolar lavage fluid (BALF) cell counts were similar in both groups, neutrophil recruitment to the lungs during RSV infection was significantly reduced in Mmp9-/- mice. Reduced neutrophil recruitment coincided with diminished RANTES, IL-1ß, SCF, G-CSF expression and p38 phosphorylation. Induction of p38 signaling was required for RANTES and G-CSF expression during RSV infection in airway epithelial cells. Therefore, MMP9 in RSV lung infection significantly enhances neutrophil recruitment, cytokine production and viral clearance while reducing AHR.


Asunto(s)
Hiperreactividad Bronquial/virología , Metaloproteinasa 9 de la Matriz/fisiología , Infecciones por Virus Sincitial Respiratorio/enzimología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitial Respiratorio Humano/patogenicidad , Animales , Apoptosis , Hiperreactividad Bronquial/inmunología , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Infiltración Neutrófila , Hipersensibilidad Respiratoria/inducido químicamente , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/virología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología
16.
Am J Physiol Lung Cell Mol Physiol ; 309(3): L205-10, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26071553

RESUMEN

Exposure to chlorine (Cl2) damages airway and alveolar epithelia resulting in acute lung injury and reactive airway hyperresponsiveness (AHR) to methacholine. However, little is known about the effect of preexisting respiratory disease on Cl2-induced lung injury. By using a murine respiratory syncytial virus (RSV) infection model, we found that preexisting RSV infection increases Cl2 (187 ppm for 30 min)-induced lung inflammation and airway AHR at 24 h after exposure (5 days after infection). RSV infection and Cl2 exposure synergistically induced oxygen desaturation and neutrophil infiltration and increased MCP-1, MIP-1ß, IL-10, IFN-γ, and RANTES concentrations in the bronchoalveolar lavage fluid (BALF). In contrast, levels of type 2 cytokines (i.e., IL-4, IL-5, IL-9, and IL-13) were not significantly affected by either RSV infection or Cl2 exposure. Cl2 exposure, but not RSV infection, induced AHR to methacholine challenge as measured by flexiVent. Moreover, preexisting RSV infection amplified BALF levels of hyaluronan (HA) and AHR. The Cl2-induced AHR was mitigated by treatment with inter-α-trypsin inhibitor antibody, which inhibits HA signaling, suggesting a mechanism of HA-mediated AHR from exacerbated oxidative injury. Our results show for the first time that preexisting RSV infection predisposes the lung to Cl2-induced injury. These data emphasize the necessity for further research on the effects of Cl2 in vulnerable populations and the development of appropriate treatments.


Asunto(s)
Lesión Pulmonar Aguda/inducido químicamente , Contaminantes Atmosféricos/toxicidad , Cloro/toxicidad , Hipersensibilidad Respiratoria/inducido químicamente , Infecciones por Virus Sincitial Respiratorio/inmunología , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/virología , Animales , Quimiocinas/metabolismo , Ácido Hialurónico/metabolismo , Masculino , Ratones Endogámicos BALB C , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/virología , Virus Sincitiales Respiratorios
17.
PLoS One ; 9(11): e112589, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25391140

RESUMEN

Low circulating levels of 25-hydroxyvitamin D [25(OH)D] are associated with chronic lung diseases such as asthma. However, it is unclear whether vitamin D is involved in disease pathogenesis or is modified by the inflammation associated with the disease process. We hypothesized that allergic inflammation decreases the level of circulating 25(OH)D and tested this using a mice model of house dust mite (HDM) induced allergic airway inflammation. Cellular influx was measured in bronchoalvelar lavage (BAL) fluid, and allergic sensitization and 25(OH)D levels were measured in serum. Exposure to HDM caused a robust inflammatory response in the lung that was enhanced by prior influenza infection. These responses were not associated with any change in circulating levels of 25(OH)D. These data suggest that alterations in circulating 25(OH)D levels induced by Th-2 driven inflammation are unlikely to explain the cross-sectional epidemiological association between vitamin D deficiency and asthma.


Asunto(s)
Inflamación/sangre , Infecciones por Orthomyxoviridae/sangre , Pyroglyphidae/inmunología , Hipersensibilidad Respiratoria/sangre , Vitamina D/análogos & derivados , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Eosinófilos/inmunología , Eosinófilos/patología , Eosinófilos/virología , Femenino , Inmunoglobulina E/sangre , Inflamación/etiología , Inflamación/inmunología , Inflamación/virología , Virus de la Influenza A/inmunología , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Macrófagos/inmunología , Macrófagos/patología , Macrófagos/virología , Ratones , Ratones Endogámicos BALB C , Neutrófilos/inmunología , Neutrófilos/patología , Neutrófilos/virología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Hipersensibilidad Respiratoria/etiología , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/virología , Células Th2/inmunología , Células Th2/patología , Células Th2/virología , Vitamina D/sangre
18.
Antiviral Res ; 110: 124-31, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25131377

RESUMEN

Respiratory syncytial virus (RSV) causes bronchiolitis in children followed by inflammation and asthma-like symptoms. The development of preventive therapy for this virus continues to pose a challenge. Fungal immunomodulatory proteins (FIPs) exhibit anti-inflammatory function. FIP-fve is an immunomodulatory protein isolated from Flammulina velutipes. To determine whether FIP-fve affects the infection or consequence of immunity of RSV, we investigated viral titers of RSV and inflammatory cytokine levels in vivo and in vitro. Oral FIP-fve decreased RSV-induced airway hyperresponsiveness (AHR), airway inflammation, and IL-6 expression in bronchoalveolar lavage fluid (BALF) of BALB/c mice. RSV replication and interleukin 6 (IL-6) levels in RSV-infected HEp-2 cells were compared before and after FIP-fve treatment. FIP-fve inhibited viral titers on plaque assay and Western blot, as well as inhibited RSV-stimulated expression of IL-6 on ELISA and RT-PCR. The results of this study suggested that FIP-fve decreases RSV replication, RSV-induced inflammation and respiratory pathogenesis. FIP-fve is a widely used, natural compound from F.velutipes that may be a safe agent for viral prevention and even therapy.


Asunto(s)
Bronquiolitis Viral/prevención & control , Proteínas Fúngicas/farmacología , Inflamación/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Virus Sincitiales Respiratorios/inmunología , Replicación Viral/efectos de los fármacos , Animales , Bronquiolitis Viral/inmunología , Líquido del Lavado Bronquioalveolar/inmunología , Línea Celular , Femenino , Proteínas Fúngicas/inmunología , Humanos , Inmunomodulación/efectos de los fármacos , Inflamación/inmunología , Interferones/metabolismo , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Transporte de Proteínas/efectos de los fármacos , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/prevención & control , Hipersensibilidad Respiratoria/virología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/crecimiento & desarrollo
19.
Clin Exp Allergy ; 44(9): 1188-99, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25039815

RESUMEN

BACKGROUND: Although morbidity and mortality rates from asthma are highest in patients > 65 years of age, the effect of older age on airway inflammation in asthma is not well established. OBJECTIVE: To investigate age-related differences in the promotion of allergic inflammation after influenza A viral respiratory infection on antigen-specific IgE production, antigen-induced airway inflammation and airway hyperresponsiveness in mice. METHODS: To accomplish this objective, the following model system was used. Young (6 week) and aged (18 months) BALB/c mice were first infected with a non-lethal dose of influenza virus A (H/HKx31). Mice were then ovalbumin (OVA)-sensitized during the acute infection (3-days post inoculation) and then chronically underwent challenge to the airways with OVA. Forty-eight hours after the final OVA challenge, airway hyperresponsiveness (AHR), bronchoalveolar fluid (BALF) cellular and cytokine profile, antigen-specific IgE and IgG1, and lung tissue inflammation were measured. RESULTS: Age-specific differences were noted on the effect of a viral infection, allergic sensitization, airway inflammation and airway hyperresponsiveness. Serum OVA-specific IgE was significantly increased in only the aged mice infected with influenza virus. Despite greater morbidity (e.g. weight loss and sickness scores) during the acute infection in the 18-month old mice that were OVA-sensitized, there was little effect on the AHR and BALF cellular differential. In contrast, BALF neutrophils and AHR increased, but eosinophils decreased in 6-week mice that were OVA-sensitized during an acute influenza infection. CONCLUSION: With increased age in a mouse model, viral infection prior to antigen sensitization affects the airway and systemic allergic response differently. These differences may reflect distinct phenotypic features of allergic inflammation in older patients with asthma.


Asunto(s)
Antígenos/inmunología , Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/virología , Factores de Edad , Animales , Asma/inmunología , Asma/metabolismo , Asma/patología , Asma/virología , Líquido del Lavado Bronquioalveolar/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Inmunoglobulina E/inmunología , Recuento de Leucocitos , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Ratones , Infecciones por Orthomyxoviridae/complicaciones , Ovalbúmina/inmunología , Hipersensibilidad Respiratoria/metabolismo , Hipersensibilidad Respiratoria/patología
20.
J Allergy Clin Immunol ; 133(6): 1720-7, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24388637

RESUMEN

BACKGROUND: ß-Agonists are used for relief and control of asthma symptoms by reversing bronchoconstriction. They might also have anti-inflammatory properties, but the underpinning mechanisms remain poorly understood. Recently, a direct interaction between formoterol and protein phosphatase 2A (PP2A) has been described in vitro. OBJECTIVE: We sought to elucidate the molecular mechanisms by which ß-agonists exert anti-inflammatory effects in allergen-driven and rhinovirus 1B-exacerbated allergic airways disease (AAD). METHODS: Mice were sensitized and then challenged with house dust mite to induce AAD while receiving treatment with salmeterol, formoterol, or salbutamol. Mice were also infected with rhinovirus 1B to exacerbate lung inflammation and therapeutically administered salmeterol, dexamethasone, or the PP2A-activating drug (S)-2-amino-4-(4-[heptyloxy]phenyl)-2-methylbutan-1-ol (AAL[S]). RESULTS: Systemic or intranasal administration of salmeterol protected against the development of allergen- and rhinovirus-induced airway hyperreactivity and decreased eosinophil recruitment to the lungs as effectively as dexamethasone. Formoterol and salbutamol also showed anti-inflammatory properties. Salmeterol, but not dexamethasone, increased PP2A activity, which reduced CCL11, CCL20, and CXCL2 expression and reduced levels of phosphorylated extracellular signal-regulated kinase 1 and active nuclear factor κB subunits in the lungs. The anti-inflammatory effect of salmeterol was blocked by targeting the catalytic subunit of PP2A with small RNA interference. Conversely, increasing PP2A activity with AAL(S) abolished rhinovirus-induced airway hyperreactivity, eosinophil influx, and CCL11, CCL20, and CXCL2 expression. Salmeterol also directly activated immunoprecipitated PP2A in vitro isolated from human airway epithelial cells. CONCLUSIONS: Salmeterol exerts anti-inflammatory effects by increasing PP2A activity in AAD and rhinovirus-induced lung inflammation, which might potentially account for some of its clinical benefits.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2/farmacología , Albuterol/análogos & derivados , Quimiotaxis/efectos de los fármacos , Quimiotaxis/inmunología , Proteína Fosfatasa 2/metabolismo , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/metabolismo , Rhinovirus/inmunología , Agonistas de Receptores Adrenérgicos beta 2/administración & dosificación , Albuterol/administración & dosificación , Albuterol/farmacología , Animales , Antígenos Dermatofagoides/efectos adversos , Modelos Animales de Enfermedad , Activación Enzimática , Eosinófilos/efectos de los fármacos , Eosinófilos/inmunología , Eosinófilos/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/virología , Masculino , Ratones , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Infecciones por Picornaviridae/inmunología , Infecciones por Picornaviridae/metabolismo , Hipersensibilidad Respiratoria/tratamiento farmacológico , Hipersensibilidad Respiratoria/virología , Xinafoato de Salmeterol
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...