Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Proc Natl Acad Sci U S A ; 108(5): 2100-5, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21252304

RESUMEN

Gastrin-releasing peptide (GRP) is synthesized by pulmonary neuroendocrine cells in inflammatory lung diseases, such as bronchopulmonary dysplasia (BPD). Many BPD infants develop asthma, a serious disorder of intermittent airway obstruction. Despite extensive research, early mechanisms of asthma remain controversial. The incidence of asthma is growing, now affecting >300 million people worldwide. To test the hypothesis that GRP mediates asthma, we used two murine models: ozone exposure for air pollution-induced airway hyperreactivity (AHR), and ovalbumin (OVA)-induced allergic airway disease. BALB/c mice were given small molecule GRP blocking agent 77427, or GRP blocking antibody 2A11, before exposure to ozone or OVA challenge. In both models, GRP blockade abrogated AHR and bronchoalveolar lavage (BAL) macrophages and granulocytes, and decreased BAL cytokines implicated in asthma, including those typically derived from Th1 (e.g., IL-2, TNFα), Th2 (e.g., IL-5, IL-13), Th17 (IL-17), macrophages (e.g., MCP-1, IL-1), and neutrophils (KC = IL-8). Dexamethasone generally had smaller effects on all parameters. Macrophages, T cells, and neutrophils express GRP receptor (GRPR). GRP blockade diminished serine phosphorylation of GRPR with ozone or OVA. Thus, GRP mediates AHR and airway inflammation in mice, suggesting that GRP blockade is promising as a broad-spectrum therapeutic approach to treat and/or prevent asthma in humans.


Asunto(s)
Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Asma/tratamiento farmacológico , Péptido Liberador de Gastrina/antagonistas & inhibidores , Animales , Líquido del Lavado Bronquioalveolar , Femenino , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C
4.
J Immunol ; 187(9): 4800-8, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21930959

RESUMEN

Inhalation of ambient ozone alters populations of lung macrophages. However, the impact of altered lung macrophage populations on the pathobiology of ozone is poorly understood. We hypothesized that subpopulations of macrophages modulate the response to ozone. We exposed C57BL/6 mice to ozone (2 ppm × 3 h) or filtered air. At 24 h after exposure, the lungs were harvested and digested and the cells underwent flow cytometry. Analysis revealed a novel macrophage subset present in ozone-exposed mice, which were distinct from resident alveolar macrophages and identified by enhanced Gr-1(+) expression [Gr-1 macrophages (Gr-1 Macs)]. Further analysis showed that Gr-1(+) Macs exhibited high expression of MARCO, CX3CR1, and NAD(P)H:quinone oxioreductase 1. Gr-1(+) Macs were present in the absence of CCR2, suggesting that they were not derived from a CCR2-dependent circulating intermediate. Using PKH26-PCL to label resident phagocytic cells, we demonstrated that Gr-1 Macs were derived from resident lung cells. This new subset was diminished in the absence of CX3CR1. Interestingly, CX3CR1-null mice exhibited enhanced responses to ozone, including increased airway hyperresponsiveness, exacerbated neutrophil influx, accumulation of 8-isoprostanes and protein carbonyls, and increased expression of cytokines (CXCL2, IL-1ß, IL-6, CCL2, and TNF-α). Our results identify a novel subset of lung macrophages, which are derived from a resident intermediate, are dependent upon CX3CR1, and appear to protect the host from the biological response to ozone.


Asunto(s)
Diferenciación Celular/inmunología , Mediadores de Inflamación/fisiología , Macrófagos Alveolares/inmunología , Estrés Oxidativo/inmunología , Ozono/administración & dosificación , Receptores de Quimiocina/fisiología , Administración Intranasal , Animales , Receptor 1 de Quimiocinas CX3C , Diferenciación Celular/genética , Femenino , Inflamación/genética , Inflamación/inmunología , Inflamación/prevención & control , Mediadores de Inflamación/administración & dosificación , Mediadores de Inflamación/uso terapéutico , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Estrés Oxidativo/genética , Ozono/efectos adversos , Receptores de Quimiocina/biosíntesis , Receptores de Quimiocina/deficiencia , Receptores de Quimiocina/genética , Receptores Inmunológicos/biosíntesis , Receptores Inmunológicos/genética
5.
Am J Respir Cell Mol Biol ; 46(4): 454-60, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22052876

RESUMEN

Air pollutant exposure is linked with childhood asthma incidence and exacerbations, and maternal exposure to airborne pollutants during pregnancy increases airway hyperreactivity (AHR) in offspring. To determine if exposure to diesel exhaust (DE) during pregnancy worsened postnatal ozone-induced AHR, timed pregnant C57BL/6 mice were exposed to DE (0.5 or 2.0 mg/m(3)) 4 hours daily from Gestation Day 9-17, or received twice-weekly oropharyngeal aspirations of the collected DE particles (DEPs). Placentas and fetal lungs were harvested on Gestation Day 18 for cytokine analysis. In other litters, pups born to dams exposed to air or DE, or to dams treated with aspirated diesel particles, were exposed to filtered air or 1 ppm ozone beginning the day after birth, for 3 hours per day, 3 days per week for 4 weeks. Additional pups were monitored after a 4-week recovery period. Diesel inhalation or aspiration during pregnancy increased levels of placental and fetal lung cytokines. There were no significant effects on airway leukocytes, but prenatal diesel augmented ozone-induced elevations of bronchoalveolar lavage cytokines at 4 weeks. Mice born to the high-concentration diesel-exposed dams had worse ozone-induced AHR, which persisted in the 4-week recovery animals. Prenatal diesel exposure combined with postnatal ozone exposure also worsened secondary alveolar crest development. We conclude that maternal inhalation of DE in pregnancy provokes a fetal inflammatory response that, combined with postnatal ozone exposure, impairs alveolar development, and causes a more severe and long-lasting AHR to ozone exposure.


Asunto(s)
Hiperreactividad Bronquial/etiología , Exposición por Inhalación , Exposición Materna , Ozono/efectos adversos , Emisiones de Vehículos/toxicidad , Contaminantes Atmosféricos/efectos adversos , Animales , Animales Recién Nacidos , Citocinas/metabolismo , Femenino , Edad Gestacional , Pulmón/efectos de los fármacos , Pulmón/embriología , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Placenta/efectos de los fármacos , Placenta/metabolismo , Neumonía/etiología , Embarazo , Alveolos Pulmonares/diagnóstico por imagen , Alveolos Pulmonares/efectos de los fármacos , Ultrasonografía
7.
J Immunol ; 185(11): 6891-8, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21037098

RESUMEN

Hyaluronan is a high-molecular mass component of pulmonary extracelluar matrix, and lung injury can generate a low-molecular mass hyaluronan (HA) fragment that functions as endogenous ligand to cell surface receptors CD44 and TLR4. This leads to activation of intracellular NF-κB signaling and proinflammatory cytokine production. Based on previous information that ozone exposure causes increased HA in bronchial alveolar lavage fluid and ozone pre-exposure primes immune response to inhaled LPS, we hypothesized that HA production during ozone exposure augments the inflammatory response to LPS. We demonstrate that acute ozone exposure at 1 part per million for 3 h primes the immune response to low-dose aerosolized LPS in C57BL/6J mice, resulting in increased neutrophil recruitment into the airspaces, increased levels of protein and proinflammatory cytokines in the bronchoalveolar lavage fluid, and increased airway hyperresponsiveness. Intratracheal instillation of endotoxin-free HA (25 µg) enhances the biological response to inhaled LPS in a manner similar to ozone pre-exposure. In vitro studies using bone marrow-derived macrophages indicate that HA enhances LPS responses measured by TNF-α production, while immunofluorescence staining of murine alveolar macrophages demonstrates that HA induces TLR4 peripheralization and lipid raft colocalization. Collectively, our observations support that ozone primes macrophage responsiveness to low-dose LPS, in part, due to HA-induced TLR4 peripheralization in lung macrophages.


Asunto(s)
Ácido Hialurónico/toxicidad , Lipopolisacáridos/administración & dosificación , Ozono/toxicidad , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Administración por Inhalación , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Células Cultivadas , Relación Dosis-Respuesta Inmunológica , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/biosíntesis , Ácido Hialurónico/química , Lipopolisacáridos/química , Lipopolisacáridos/farmacología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Peso Molecular , Ozono/administración & dosificación , Ozono/química
9.
Am J Respir Crit Care Med ; 183(12): 1644-52, 2011 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-21471107

RESUMEN

RATIONALE: Previously, we demonstrated a candidate region for susceptibility to airspace enlargement on mouse chromosome 5. However, the specific candidate genes within this region accounting for emphysema-like changes remain unrecognized. c-Kit is a receptor tyrosine kinase within this candidate gene region that has previously been recognized to contribute to the survival, proliferation, and differentiation of hematopoietic stem cells. Increases in the percentage of cells expressing c-Kit have previously been associated with protection against injury-induced emphysema. OBJECTIVES: Determine whether genetic variants of c-Kit are associated with spontaneous airspace enlargement. METHODS: Perform single-nucleotide polymorphism association studies in the mouse strains at the extremes of airspace enlargement phenotype for variants in c-Kit tyrosine kinase. Characterize mice bearing functional variants of c-Kit compared with wild-type controls for the development of spontaneous airspace enlargement. Epithelial cell proliferation was measured in culture. MEASUREMENTS AND MAIN RESULTS: Upstream regulatory single-nucleotide polymorphisms in the divergent mouse strains were associated with the lung compliance difference observed between the extreme strains. c-Kit mutant mice (Kit(W-sh)/(W-sh)), when compared with genetic controls, developed altered lung histology, increased total lung capacity, increased residual volume, and increased lung compliance that persist into adulthood. c-Kit inhibition with imatinib attenuated in vitro proliferation of cells expressing epithelial cell adhesion molecule. CONCLUSIONS: Our findings indicate that c-Kit sustains and/or maintains normal alveolar architecture in the lungs of mice. In vitro data suggest that c-Kit can regulate epithelial cell clonal expansion. The precise mechanisms that c-Kit contributes to the development of airspace enlargement and increased lung compliance remain unclear and warrants further investigation.


Asunto(s)
Enfisema/prevención & control , Proteínas Proto-Oncogénicas c-kit/fisiología , Alveolos Pulmonares/fisiología , Animales , Enfisema/patología , Predisposición Genética a la Enfermedad , Pulmón/fisiopatología , Rendimiento Pulmonar/fisiología , Ratones , Ratones Endogámicos/fisiología , Ratones Mutantes , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas c-kit/genética , Alveolos Pulmonares/citología
10.
Proc Natl Acad Sci U S A ; 106(28): 11691-6, 2009 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-19581591

RESUMEN

Trachealess (Trh) is a PAS domain transcription factor regulating Drosophila tracheogenesis. No other Trh homolog has been associated with a respiratory phenotype. Seeking homolog(s) regulating lung development, we screened murine genomic DNA using trh oligonucleotides, identifying only Npas3. Npas3 mRNA peaks in lung from E10.5 to E13.5, verified by sequencing, with immunostaining in airway epithelial cells. Npas3-null mice have reduced lung branching morphogenesis but are viable prenatally. Npas3-null newborns die in respiratory distress, with diminished alveolarization, decreased Shh, Fgf9, Fgf10, and Bmp4 mRNAs, and increased Spry2, consistent with reduced FGF signaling. Exogenous FGF10 rescues branching morphogenesis in Npas3-null lungs. In promoter reporter assays, NPAS3 directly upregulates Shh and represses Spry2. Npas3(+/-) mice have a milder lung phenotype, surviving postnatally, but develop emphysema and airways hyperreactivity. Therefore, absence of a developmentally expressed transcription factor can alter downstream gene expression and multiple signaling pathways in organogenesis. NPAS3 haploinsufficiency may also lead to emphysema and asthma.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Pulmón/metabolismo , Morfogénesis/genética , Animales , Secuencia de Bases , Western Blotting , Proteínas de Drosophila/genética , Perfilación de la Expresión Génica , Inmunohistoquímica , Pulmón/crecimiento & desarrollo , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Oligonucleótidos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Factores de Transcripción/genética
11.
Am J Physiol Lung Cell Mol Physiol ; 301(5): L739-44, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21784966

RESUMEN

S-nitrosoglutathione (GSNO) is an endogenous bronchodilator present in micromolar concentrations in airway lining fluid. Airway GSNO levels decrease in severe respiratory failure and asthma, which is attributable to increased metabolism by GSNO reductase (GSNOR). Indeed, we have found that GSNOR expression and activity correlate inversely with lung S-nitrosothiol (SNO) content and airway hyperresponsiveness (AHR) to methacholine (MCh) challenge in humans with asthmatic phenotypes (Que LG, Yang Z, Stamler JS, Lugogo NL, Kraft M. Am J Respir Crit Care Med 180: 226-231, 2009). Accordingly, we hypothesized that local aerosol delivery of GSNO could ameliorate AHR and inflammation in the ovalbumin-sensitized and -challenged (OVA) mouse model of allergic asthma. Anesthetized, paralyzed, and tracheotomized 6-wk-old male control and OVA C57BL/6 mice were administered a single 15-s treatment of 0-100 mM GSNO. Five minutes later, airway resistance to MCh was measured and SNOs were quantified in bronchoalveolar lavage (BAL). Duration of protection was evaluated following nose-only exposure to 10 mM GSNO for 10 min followed by measurements of airway resistance, inflammatory cells, and cytokines and chemokines at up to 4 h later. Acute delivery of GSNO aerosol protected OVA mice from MCh-induced AHR, with no benefit seen above 20 mM GSNO. The antibronchoconstrictive effects of GSNO aerosol delivered via nose cone were sustained for at least 4 h. However, administration of GSNO did not alter total BAL cell counts or cell differentials and had modest effects on cytokine and chemokine levels. In conclusion, in the OVA mouse model of allergic asthma, aerosolized GSNO has rapid and sustained antibronchoconstrictive effects but does not substantially alter airway inflammation.


Asunto(s)
Hiperreactividad Bronquial/inducido químicamente , Hiperreactividad Bronquial/tratamiento farmacológico , Broncoconstricción/efectos de los fármacos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Pulmón/efectos de los fármacos , S-Nitrosoglutatión , Administración por Inhalación , Aerosoles/administración & dosificación , Animales , Hiperreactividad Bronquial/metabolismo , Hiperreactividad Bronquial/patología , Hiperreactividad Bronquial/fisiopatología , Pruebas de Provocación Bronquial , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Citocinas/análisis , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Inflamación/metabolismo , Inflamación/patología , Inflamación/fisiopatología , Pulmón/metabolismo , Pulmón/patología , Pulmón/fisiopatología , Masculino , Cloruro de Metacolina/efectos adversos , Ratones , Ratones Endogámicos C57BL , Mucina 5AC/análisis , Mucina 5AC/biosíntesis , Ovalbúmina/efectos adversos , S-Nitrosoglutatión/administración & dosificación , S-Nitrosoglutatión/uso terapéutico , Traqueotomía
12.
Am J Physiol Lung Cell Mol Physiol ; 301(3): L327-33, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21724860

RESUMEN

Inducible nitric oxide synthase (NOS2) expression is increased in the airway epithelium in acute inflammatory disorders although the physiological impact remains unclear. We have previously shown that NOS2 inhibits NF-κB (p50-p65) activation in respiratory epithelial cells by inducing S-nitrosylation of the p65 monomer (SNO-p65). In addition, we have demonstrated that mouse lung SNO-p65 levels are acutely depleted in a lipopolysaccharide (LPS) model of lung injury and that augmenting SNO-p65 levels before LPS treatment results in decreased airway epithelial NF-κB activation, airway inflammation, and lung injury. We now show that aerosolized LPS induces NOS2 expression in the respiratory epithelium concomitant with an increase in lung SNO-p65 levels and a decrease in airway NF-κB activity. Genetic deletion of NOS2 results in an absence of SNO-p65 formation, persistent NF-κB activity in the respiratory epithelium, and prolonged airway inflammation. These results indicate that a primary function of LPS-induced NOS2 expression in the respiratory epithelium is to modulate the inflammatory response through deactivation of NF-κB via S-nitrosylation of p65, thereby counteracting the initial stimulus-coupled denitrosylation.


Asunto(s)
Óxido Nítrico Sintasa de Tipo II/fisiología , Mucosa Respiratoria/metabolismo , Factor de Transcripción ReIA/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/química , Inflamación/inducido químicamente , Lipopolisacáridos , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/biosíntesis
13.
J Clin Invest ; 118(10): 3462-9, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18802477

RESUMEN

Asthma is a complex heritable disease that is increasing in prevalence and severity, particularly in developed countries such as the United States, where 11% of the population is affected. The contribution of environmental and genetic factors to this growing epidemic is currently not well understood. We developed the hypothesis, based on previous literature, that changes in DNA methylation resulting in aberrant gene transcription may enhance the risk of developing allergic airway disease. Our findings indicate that in mice, a maternal diet supplemented with methyl donors enhanced the severity of allergic airway disease that was inherited transgenerationally. Using a genomic approach, we discovered 82 gene-associated loci that were differentially methylated after in utero supplementation with a methyl-rich diet. These methylation changes were associated with decreased transcriptional activity and increased disease severity. Runt-related transcription factor 3 (Runx3), a gene known to negatively regulate allergic airway disease, was found to be excessively methylated, and Runx3 mRNA and protein levels were suppressed in progeny exposed in utero to a high-methylation diet. Moreover, treatment with a demethylating agent increased Runx3 gene transcription, further supporting our claim that a methyl-rich diet can affect methylation status and consequent transcriptional regulation. Our findings indicate that dietary factors can modify the heritable risk of allergic airway disease through epigenetic mechanisms during a vulnerable period of fetal development in mice.


Asunto(s)
Asma/metabolismo , Metilación de ADN , Suplementos Dietéticos , Modelos Animales de Enfermedad , Efectos Tardíos de la Exposición Prenatal , Animales , Azacitidina/farmacología , Citocinas/metabolismo , Metilación de ADN/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Epigénesis Genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Linfocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Embarazo , Fenómenos Fisiologicos de la Nutrición Prenatal , Reproducibilidad de los Resultados , Bazo/citología , Bazo/metabolismo
14.
J Immunol ; 182(12): 7818-27, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19494306

RESUMEN

The lung is constantly challenged during normal breathing by a myriad of environmental irritants and infectious insults. Pulmonary host defense mechanisms maintain homeostasis between inhibition/clearance of pathogens and regulation of inflammatory responses that could injure the airway epithelium. One component of this defense mechanism, surfactant protein-A (SP-A), exerts multifunctional roles in mediating host responses to inflammatory and infectious agents. SP-A has a bacteriostatic effect on Mycoplasma pneumoniae (Mp), which occurs by binding surface disaturated phosphatidylglycerols. SP-A can also bind the Mp membrane protein, MPN372. In this study, we investigated the role of SP-A during acute phase pulmonary infection with Mp using mice deficient in SP-A. Biologic responses, inflammation, and cellular infiltration, were much greater in Mp infected SP-A(-/-) mice than wild-type mice. Likewise, physiologic responses (airway hyperresponsiveness and lung compliance) to Mp infection were more severely affected in SP-A(-/-) mice. Both Mp-induced biologic and physiologic changes were attenuated by pharmacologic inhibition of TNF-alpha. Our findings demonstrate that SP-A is vital to preserving lung homeostasis and host defense to this clinically relevant strain of Mp by curtailing inflammatory cell recruitment and limiting an overzealous TNF-alpha response.


Asunto(s)
Homeostasis/inmunología , Mycoplasma pneumoniae/fisiología , Neumonía por Mycoplasma/inmunología , Neumonía por Mycoplasma/patología , Proteína A Asociada a Surfactante Pulmonar/metabolismo , Animales , Lavado Broncoalveolar , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía por Mycoplasma/genética , Neumonía por Mycoplasma/metabolismo , Proteína A Asociada a Surfactante Pulmonar/deficiencia , Proteína A Asociada a Surfactante Pulmonar/genética , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/inmunología
15.
Am J Respir Crit Care Med ; 181(7): 666-75, 2010 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-20007931

RESUMEN

RATIONALE: Ozone is a common environmental air pollutant that contributes to hospitalizations for respiratory illness. The mechanisms, which regulate ozone-induced airway hyperresponsiveness, remain poorly understood. We have previously reported that toll-like receptor 4 (TLR4)-deficient animals are protected against ozone-induced airway hyperresponsiveness (AHR) and that hyaluronan (HA) mediates ozone-induced AHR. However, the relation between TLR4 and hyaluronan in the airway response to ozone remains unexplored. OBJECTIVES: We hypothesized that HA acts as an endogenous TLR4 ligand for the development of AHR after ozone-induced environmental airway injury. METHODS: TLR4-deficient and wild-type C57BL/6 mice were exposed to either inhaled ozone or intratracheal HA and the inflammatory and AHR response was measured. MEASUREMENTS AND MAIN RESULTS: TLR4-deficient mice have similar levels of cellular inflammation, lung injury, and soluble HA levels as those of C57BL/6 mice after inhaled ozone exposure. However, TLR4-deficient mice are partially protected from AHR after ozone exposure as well as after direct intratracheal instillation of endotoxin-free low molecular weight HA. Similar patterns of TLR4-dependent cytokines were observed in the bronchial alveolar lavage fluid after exposure to either ozone or HA. Exposure to ozone increased immunohistological staining of TLR4 on lung macrophages. Furthermore, in vitro HA exposure of bone marrow-derived macrophages induced NF-kappaB and production of a similar pattern of proinflammatory cytokines in a manner dependent on TLR4. CONCLUSIONS: Our observations support the observation that extracellular matrix HA contributes to ozone-induced airways disease. Furthermore, our results support that TLR4 contributes to the biological response to HA by mediating both the production of proinflammatory cytokines and the development of ozone-induced AHR.


Asunto(s)
Hiperreactividad Bronquial/inducido químicamente , Hiperreactividad Bronquial/metabolismo , Ácido Hialurónico/metabolismo , Ozono/administración & dosificación , Receptor Toll-Like 4/metabolismo , Administración por Inhalación , Animales , Hiperreactividad Bronquial/patología , Líquido del Lavado Bronquioalveolar/química , Citocinas/biosíntesis , Ácido Hialurónico/administración & dosificación , Mediadores de Inflamación/metabolismo , Macrófagos Alveolares/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ozono/toxicidad , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Receptor Toll-Like 4/deficiencia
16.
Am J Physiol Lung Cell Mol Physiol ; 299(3): L345-52, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20543006

RESUMEN

In a mouse model of neutrophil elastase-induced bronchitis that exhibits goblet cell metaplasia and inflammation, we investigated the effects of intratracheal instillation of the MANS peptide, a peptide identical to the NH(2) terminus of the myristoylated alanine-rich C kinase substrate (MARCKS) on mucin protein airway secretion, inflammation, and airway reactivity. To induce mucus cell metaplasia in the airways, male BALB/c mice were treated repetitively with the serine protease, neutrophil elastase, on days 1, 4, and 7. On day 11, when goblet cell metaplasia was fully developed and profiles of proinflammatory cytokines were maximal, the animals were exposed to aerosolized methacholine after intratracheal instillation of MANS or a missense control peptide (RNS). MANS, but not RNS, attenuated the methacholine-stimulated secretion of the major respiratory mucin protein, Muc5ac (50% reduction). Concurrently, elastase-induced proinflammatory cytokines typically recovered in bronchoalveolar lavage (BAL), including KC, IL-1beta, IL-6, MCP-1, and TNFalpha, were reduced by the MANS peptide (mean levels decreased 50-60%). Secondary to the effects of MANS on mucin secretion and inflammation, mechanical lung function by forced oscillation technique was characterized with respect to airway reactivity in response to cumulative aerosol stimulation with serotonin. The MANS peptide was also found to effectively attenuate airway hyperresponsiveness to serotonin in this airway hypersecretory model. Collectively, these findings support the concept that even in airway epithelia remodeled with goblet cell metaplasia and in a state of mucin hypersecretion, exogenous attenuation of function of MARCKS protein via the MANS peptide decreases airway mucin secretion, inflammation, and hyperreactivity.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Proteínas de la Membrana/antagonistas & inhibidores , Mucina 5AC/metabolismo , Fragmentos de Péptidos/farmacología , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/metabolismo , Aerosoles , Animales , Líquido del Lavado Bronquioalveolar/química , Citocinas/metabolismo , Células Caliciformes/patología , Inflamación/prevención & control , Mediadores de Inflamación/metabolismo , Elastasa de Leucocito/farmacología , Pulmón/efectos de los fármacos , Pulmón/fisiopatología , Masculino , Metaplasia/inducido químicamente , Cloruro de Metacolina/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Mucina 5AC/antagonistas & inhibidores , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada , Hipersensibilidad Respiratoria/inducido químicamente , Hipersensibilidad Respiratoria/fisiopatología , Serotonina
18.
Pediatr Res ; 68(1): 70-4, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20386143

RESUMEN

Premature infants are at increased risk of developing airway hyper-reactivity (AHR) after oxidative stress and inflammation. Mast cells contribute to AHR partly by mediator release, so we sought to determine whether blocking mast cell degranulation or recruitment prevents hyperoxia-induced AHR, mast cell accumulation, and airway smooth muscle (ASM) changes. Rats were exposed at birth to air or 60% O2 for 14 d, inducing significantly increased AHR in the latter group, induced by nebulized methacholine challenge and measured by forced oscillometry. Daily treatment (postnatal d 1-14) with intraperitoneal cromolyn prevented hyperoxia-induced AHR, as did treatment with imatinib on postnatal d 5-14, compared with vehicle treated controls. Cromolyn prevented mast cell degranulation in the trachea but not hilar airways and blocked mast cell accumulation in the hilar airways. Imatinib treatment completely blocked mast cell accumulation in tracheal/hilar airway tissues. Hyperoxia-induced AHR in neonatal rats is mediated, at least in part, via the mast cell.


Asunto(s)
Hiperreactividad Bronquial/fisiopatología , Hiperoxia/metabolismo , Mastocitos/fisiología , Animales , Animales Recién Nacidos , Antiasmáticos/farmacología , Benzamidas , Broncoconstrictores/farmacología , Cromolin Sódico/farmacología , Femenino , Mesilato de Imatinib , Pulmón/citología , Pulmón/efectos de los fármacos , Pulmón/fisiología , Mastocitos/efectos de los fármacos , Cloruro de Metacolina/farmacología , Piperazinas/farmacología , Embarazo , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Respiración
19.
J Leukoc Biol ; 85(1): 124-31, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18818374

RESUMEN

Asthma remains a major cause of morbidity and hospitalizations in developed nations. Despite the widespread prevalence of this disease, the genetic and environmental factors that mediate development and progression of allergic airways disease remain poorly understood. Pulmonary recruitment of eosinophils is believed to contribute to many cardinal features of allergic airways disease. Therefore, it is paramount to understand host factors that contribute to pulmonary eosinophil recruitment into the lungs. Mindin is a component of pulmonary extracellular matrix, which can regulate inflammatory cell recruitment. We characterized the role of mindin in the severity of allergic airways disease using established murine models. There were no baseline differences in wild-type and mindin-deficient animals in cell counts or airway physiology. Using the OVA murine model of allergic airways disease, we observed that mindin-deficient animals have less-severe allergic airways disease with fewer airspace eosinophils and lower lung-lavage levels of inflammatory Th2 cytokines such as IL-13 and IL-4. Furthermore, mindin-deficient animals have reduced airway hyper-responsiveness after methacholine challenge. To determine the role of mindin in eosinophil trafficking, independent of antigen immunization or T lymphocyte activation, we instilled IL-13 directly into the lungs of mice. In this model, mindin regulates eosinophil recruitment into the airspace. In vitro experiments demonstrate that mindin can enhance eotaxin-mediated eosinophil adhesion and migration, which are dependent on the expression of integrins alphaMbeta2 and alpha4beta1. In conclusion, these data suggest that mindin participates in integrin-dependent trafficking of eosinophils and can contribute to the severity of allergic airways disease.


Asunto(s)
Movimiento Celular , Eosinófilos/fisiología , Proteínas de la Matriz Extracelular/fisiología , Hipersensibilidad/patología , Pulmón/patología , Animales , Asma/inmunología , Asma/patología , Adhesión Celular/fisiología , Proteínas de la Matriz Extracelular/genética , Hipersensibilidad/inmunología , Inmunoglobulina E/metabolismo , Técnicas In Vitro , Integrina alfa4beta1/metabolismo , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Pulmón/efectos de los fármacos , Pulmón/inmunología , Antígeno de Macrófago-1/metabolismo , Cloruro de Metacolina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovalbúmina/inmunología
20.
Am J Respir Crit Care Med ; 180(12): 1218-26, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19762564

RESUMEN

RATIONALE: Epidemiologic studies implicate air pollutant exposure during pregnancy as a risk factor for wheezing in offspring. Ozone exposure is linked to exacerbations of wheezing in children. OBJECTIVES: To determine if maternal pulmonary exposure to traffic-related particles during pregnancy augments ozone-induced airway hyperresponsiveness in offspring. METHODS: C57BL6 time-mated mice were given NIST SRM#1648 (particulate matter [PM]) 0.48 mg, saline vehicle, or no treatment by tracheal insufflation twice weekly for 3 weeks. PM exposure augmented maternal lung inflammation and placental TNF-alpha, Keratinocyte-derived cytokine (KC), and IL-6 (measured at gestation Day 18). After parturition, dams and litters were exposed to air or ozone 1 ppm 3 h/d, every other day, thrice weekly for 4 weeks. Respiratory system resistance in pups was measured at baseline and after administration of nebulized methacholine. MEASUREMENTS AND MAIN RESULTS: Ozone increased airway hyperresponsiveness, but the increase was greatest in pups born to PM-treated dams. Whole-lung TNF-alpha, IL-1beta, KC, IL-6, and MCP-1 were increased in ozone-treated pups, with the greatest increase in pups born to dams given PM. Airway epithelial mucous metaplasia estimated by periodic acid-Schiff Alcian blue staining was increased in ozone-exposed pups born to PM-treated dams. Alveolar development, determined by morphometry, and airway smooth muscle bulk, estimated using alpha-actin histochemistry, were unaffected by prenatal or postnatal treatment. CONCLUSIONS: Maternal pulmonary exposure to PM during pregnancy augments placental cytokine expression and postnatal ozone-induced pulmonary inflammatory cytokine responses and ozone-induced airway hyperresponsiveness without altering airway structure.


Asunto(s)
Hiperreactividad Bronquial/inducido químicamente , Exposición Materna , Oxidantes Fotoquímicos/toxicidad , Ozono/toxicidad , Material Particulado/toxicidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Contaminantes Atmosféricos/toxicidad , Contaminación del Aire , Análisis de Varianza , Animales , Hiperreactividad Bronquial/fisiopatología , Pruebas de Provocación Bronquial , Líquido del Lavado Bronquioalveolar , Modelos Animales de Enfermedad , Femenino , Inflamación/inducido químicamente , Inflamación/fisiopatología , Exposición por Inhalación , Pulmón/fisiopatología , Intercambio Materno-Fetal , Ratones , Ratones Endogámicos C57BL , Embarazo , Pruebas de Función Respiratoria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA