Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Exp Allergy ; 48(6): 641-649, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29383778

RESUMEN

BACKGROUND: Adipose tissue-derived inflammation is linked to obesity-related comorbidities. This study aimed to quantify and immuno-phenotype adipose tissue macrophages (ATMs) from obese asthmatics and obese non-asthmatics and to examine associations between adipose tissue, systemic and airway inflammation. METHODS: Visceral (VAT) adipose tissue and subcutaneous (SAT) adipose tissue were collected from obese adults undergoing bariatric surgery and processed to obtain the stromovascular fraction. Pro-inflammatory (M1) and anti-inflammatory (M2) macrophages were quantified by flow cytometry. Cytospins of induced sputum were stained for differential cell counts. Plasma C-reactive protein (CRP) and CD163 were measured by ELISA. RESULTS: VAT contained a higher number of ATMs compared to SAT. A higher percentage of M1 ATMs was observed in VAT of obese asthmatics compared to obese non-asthmatics. The M1:M2 ratio in VAT was negatively associated with FEV1 %. Sputum macrophage count was correlated positively with M1 ATMs and negatively with M2 ATMs in VAT. In obese asthmatics, CRP was positively associated with M1:M2 ratio in VAT. There were no associations with CD163. An elevated ratio of M1:M2 ATMs was observed in VAT of obese asthmatics with increased disease severity. CONCLUSIONS AND CLINICAL RELEVANCE: Visceral inflammation with increased pro-inflammatory macrophages (M1) occurs in obese asthma and may be a determinant of systemic inflammation and asthma severity.


Asunto(s)
Tejido Adiposo/inmunología , Asma/diagnóstico , Asma/etiología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Obesidad/complicaciones , Tejido Adiposo/patología , Adulto , Biomarcadores , Composición Corporal , Estudios Transversales , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Inflamación/metabolismo , Inflamación/patología , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad , Fenotipo , Pruebas de Función Respiratoria
2.
Allergy ; 72(12): 1891-1903, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28543283

RESUMEN

BACKGROUND: Asthma is an allergic airway disease (AAD) caused by aberrant immune responses to allergens. Protein phosphatase-2A (PP2A) is an abundant serine/threonine phosphatase with anti-inflammatory activity. The ubiquitin proteasome system (UPS) controls many cellular processes, including the initiation of inflammatory responses by protein degradation. We assessed whether enhancing PP2A activity with fingolimod (FTY720) or 2-amino-4-(4-(heptyloxy) phenyl)-2-methylbutan-1-ol (AAL(S) ), or inhibiting proteasome activity with bortezomib (BORT), could suppress experimental AAD. METHODS: Acute AAD was induced in C57BL/6 mice by intraperitoneal sensitization with ovalbumin (OVA) in combination with intranasal (i.n) exposure to OVA. Chronic AAD was induced in mice with prolonged i.n exposure to crude house dust mite (HDM) extract. Mice were treated with vehicle, FTY720, AAL(S) , BORT or AAL(S) +BORT and hallmark features of AAD assessed. RESULTS: AAL(S) reduced the severity of acute AAD by suppressing tissue eosinophils and inflammation, mucus-secreting cell (MSC) numbers, type 2-associated cytokines (interleukin (IL)-33, thymic stromal lymphopoietin, IL-5 and IL-13), serum immunoglobulin (Ig)E and airway hyper-responsiveness (AHR). FTY720 only suppressed tissue inflammation and IgE. BORT reduced bronchoalveolar lavage fluid (BALF) and tissue eosinophils and inflammation, IL-5, IL-13 and AHR. Combined treatment with AAL(S) +BORT had complementary effects and suppressed BALF and tissue eosinophils and inflammation, MSC numbers, reduced the production of type 2 cytokines and AHR. AAL(S) , BORT and AAL(S) +BORT also reduced airway remodelling in chronic AAD. CONCLUSION: These findings highlight the potential of combination therapies that enhance PP2A and inhibit proteasome activity as novel therapeutic strategies for asthma.


Asunto(s)
Antiasmáticos/farmacología , Inhibidores Enzimáticos/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Proteína Fosfatasa 2/antagonistas & inhibidores , Hipersensibilidad Respiratoria/etiología , Hipersensibilidad Respiratoria/metabolismo , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Biomarcadores , Citocinas , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Expresión Génica , Mediadores de Inflamación/metabolismo , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Hipersensibilidad Respiratoria/tratamiento farmacológico , Hipersensibilidad Respiratoria/patología
3.
Mucosal Immunol ; 9(4): 859-72, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26555706

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a life-threatening inflammatory respiratory disorder, often induced by cigarette smoke (CS) exposure. The development of effective therapies is impaired by a lack of understanding of the underlining mechanisms. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a cytokine with inflammatory and apoptotic properties. We interrogated a mouse model of CS-induced experimental COPD and human tissues to identify a novel role for TRAIL in COPD pathogenesis. CS exposure of wild-type mice increased TRAIL and its receptor messenger RNA (mRNA) expression and protein levels, as well as the number of TRAIL(+)CD11b(+) monocytes in the lung. TRAIL and its receptor mRNA were also increased in human COPD. CS-exposed TRAIL-deficient mice had decreased pulmonary inflammation, pro-inflammatory mediators, emphysema-like alveolar enlargement, and improved lung function. TRAIL-deficient mice also developed spontaneous small airway changes with increased epithelial cell thickness and collagen deposition, independent of CS exposure. Importantly, therapeutic neutralization of TRAIL, after the establishment of early-stage experimental COPD, reduced pulmonary inflammation, emphysema-like alveolar enlargement, and small airway changes. These data provide further evidence for TRAIL being a pivotal inflammatory factor in respiratory diseases, and the first preclinical evidence to suggest that therapeutic agents that target TRAIL may be effective in COPD therapy.


Asunto(s)
Inflamación/inmunología , Pulmón/inmunología , Monocitos/inmunología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , ARN Mensajero/genética , Mucosa Respiratoria/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Animales , Apoptosis , Modelos Animales de Enfermedad , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Fumar/efectos adversos , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Regulación hacia Arriba
4.
Mucosal Immunol ; 9(3): 809-20, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26509876

RESUMEN

Exposure to particulate matter (PM), a major component of air pollution, contributes to increased morbidity and mortality worldwide. PM induces innate immune responses and contributes to allergic sensitization, although the mechanisms governing this process remain unclear. Lung mucosal uric acid has also been linked to allergic sensitization. The links among PM exposure, uric acid, and allergic sensitization remain unexplored. We therefore investigated the mechanisms behind PM-induced allergic sensitization in the context of lung mucosal uric acid. PM10 and house dust mite exposure selectively induced lung mucosal uric acid production and secretion in vivo, which did not occur with other challenges (lipopolysaccharide, virus, bacteria, or inflammatory/fibrotic stimuli). PM10-induced uric acid mediates allergic sensitization and augments antigen-specific T-cell proliferation, which is inhibited by uricase. We then demonstrate that human airway epithelial cells secrete uric acid basally and after stimulation through a previously unidentified mucosal secretion system. Our work discovers a previously unknown mechanism of air pollution-induced, uric acid-mediated, allergic sensitization that may be important in the pathogenesis of asthma.


Asunto(s)
Antígenos Dermatofagoides/inmunología , Hipersensibilidad/inmunología , Pulmón/fisiología , Material Particulado/inmunología , Mucosa Respiratoria/inmunología , Linfocitos T/inmunología , Ácido Úrico/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Exposición a Riesgos Ambientales/efectos adversos , Femenino , Humanos , Inmunidad Mucosa , Inmunización , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pyroglyphidae , Mucosa Respiratoria/patología , Receptor Toll-Like 4/genética
5.
Mucosal Immunol ; 7(3): 478-88, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24045576

RESUMEN

Respiratory infections in early life can lead to chronic respiratory disease. Chlamydia infections are common causes of respiratory disease, particularly pneumonia in neonates, and are linked to permanent reductions in pulmonary function and the induction of asthma. However, the immune responses that protect against early-life infection and the mechanisms that lead to chronic lung disease are incompletely understood. Here we identify novel roles for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in promoting Chlamydia respiratory infection-induced pathology in early life, and subsequent chronic lung disease. By infecting TRAIL-deficient neonatal mice and using neutralizing antibodies against this factor and its receptors in wild-type mice, we demonstrate that TRAIL is critical in promoting infection-induced histopathology, inflammation, and mucus hypersecretion, as well as subsequent alveolar enlargement and impaired lung function. This suggests that therapeutic agents that target TRAIL or its receptors may be effective treatments for early-life respiratory infections and associated chronic lung disease.


Asunto(s)
Neumonía/metabolismo , Infecciones del Sistema Respiratorio/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Animales , Animales Recién Nacidos , Anticuerpos Neutralizantes/farmacología , Apoptosis/genética , Infecciones por Chlamydia/metabolismo , Chlamydia muridarum , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Expresión Génica , Ratones , Ratones Noqueados , Moco/metabolismo , FN-kappa B/metabolismo , Neumonía/genética , Neumonía/microbiología , Neumonía/patología , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/antagonistas & inhibidores , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Hipersensibilidad Respiratoria/genética , Hipersensibilidad Respiratoria/metabolismo , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/microbiología , Ligando Inductor de Apoptosis Relacionado con TNF/deficiencia , Ligando Inductor de Apoptosis Relacionado con TNF/genética
6.
Mucosal Immunol ; 6(3): 569-79, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23131786

RESUMEN

Deleterious responses to pathogens during infancy may contribute to infection and associated asthma. Chlamydia respiratory infections in early life are common causes of pneumonia and lead to reduced lung function and asthma. We investigated the role of interleukin-13 (IL-13) in promoting early-life Chlamydia respiratory infection, infection-induced airway hyperresponsiveness (AHR), and severe allergic airway disease (AAD). Infected infant Il13(-/-) mice had reduced infection, inflammation, and mucus-secreting cell hyperplasia. Surprisingly, infection of wild-type (WT) mice did not increase IL-13 production but reduced IL-13Rα2 decoy receptor levels compared with sham-inoculated controls. Infection of WT but not Il13(-/-) mice induced persistent AHR. Infection and associated pathology were restored in infected Il13(-/-) mice by reconstitution with IL-13. Stat6(-/-) mice were also largely protected. Neutralization of IL-13 during infection prevented subsequent infection-induced severe AAD. Thus, early-life Chlamydia respiratory infection reduces IL-13Rα2 production, which may enhance the effects of constitutive IL-13 and promote more severe infection, persistent AHR, and AAD.


Asunto(s)
Chlamydia/inmunología , Neumonía por Clamidia/inmunología , Interleucina-13/metabolismo , Hipersensibilidad Respiratoria/inmunología , Edad de Inicio , Animales , Animales Recién Nacidos , Anticuerpos Bloqueadores/metabolismo , Células Cultivadas , Neumonía por Clamidia/epidemiología , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Humanos , Interleucina-13/genética , Interleucina-13/inmunología , Subunidad alfa1 del Receptor de Interleucina-13/genética , Subunidad alfa1 del Receptor de Interleucina-13/inmunología , Subunidad alfa1 del Receptor de Interleucina-13/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Hipersensibilidad Respiratoria/epidemiología , Factor de Transcripción STAT6/genética
7.
J Dev Orig Health Dis ; 3(3): 153-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25102006

RESUMEN

Many important human diseases, such as asthma, have their developmental origins in early life. Respiratory infections in particular may alter the course of asthma and may either protect against or promote the development of this disease. It is likely that the nature of the effects depends on the type and age of infection and is determined by the impact of infection on the immune and respiratory systems. Immunity in early life is plastic and can be moulded by antigen encounter, which may enhance or reinforce the asthmatic phenotype of early life, or induce protective responses. Chlamydial respiratory infections have specific effects and may increase asthma severity in early life by promoting systemic interleukin 13 responses and causing permanent changes in lung structure. Respiratory viral infections, such as those of respiratory syncytial virus and rhinovirus, promote pro-asthmatic responses in early life that contribute to the induction of asthma. By contrast, probiotics or infection or exposure to certain bacteria, such as Streptococcus pneumoniae, may have protective effects in asthma by increasing the numbers and activity of regulatory T cells. Here, we review the impact of infections on the developmental origins of asthma. Understanding these effects may lead to new therapeutic approaches for asthma that either target deleterious infections or utilize beneficial ones.

8.
Eur Respir J ; 37(1): 53-64, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20525707

RESUMEN

An inverse association exists between some bacterial infections and the prevalence of asthma. We investigated whether Streptococcus pneumoniae infection protects against asthma using mouse models of ovalbumin (OVA)-induced allergic airway disease (AAD). Mice were intratracheally infected or treated with killed S. pneumoniae before, during or after OVA sensitisation and subsequent challenge. The effects of S. pneumoniae on AAD were assessed. Infection or treatment with killed S. pneumoniae suppressed hallmark features of AAD, including antigen-specific T-helper cell (Th) type 2 cytokine and antibody responses, peripheral and pulmonary eosinophil accumulation, goblet cell hyperplasia, and airway hyperresponsiveness. The effect of infection on the development of specific features of AAD depended on the timing of infection relative to allergic sensitisation and challenge. Infection induced significant increases in regulatory T-cell (Treg) numbers in lymph nodes, which correlated with the degree of suppression of AAD. Tregs reduced T-cell proliferation and Th2 cytokine release. The suppressive effects of infection were reversed by anti-CD25 treatment. Respiratory infection or treatment with S. pneumoniae attenuates allergic immune responses and suppresses AAD. These effects may be mediated by S. pneumoniae-induced Tregs. This identifies the potential for the development of therapeutic agents for asthma from S. pneumoniae.


Asunto(s)
Asma/microbiología , Hipersensibilidad/microbiología , Infecciones Estreptocócicas/metabolismo , Infecciones Estreptocócicas/parasitología , Streptococcus pneumoniae/metabolismo , Linfocitos T/microbiología , Animales , Hiperreactividad Bronquial/inmunología , Humanos , Sistema Inmunológico , Inflamación , Subunidad alfa del Receptor de Interleucina-2/biosíntesis , Pulmón/microbiología , Ratones , Ratones Endogámicos BALB C , Hipersensibilidad Respiratoria/inmunología , Linfocitos T Reguladores/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...