Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-38179428

RESUMEN

Introduction: There is a need to better understand the etiotypes of chronic obstructive pulmonary disease (COPD) beyond the tobacco-smoke (TS-COPD). Wood smoke COPD (WS-COPD) is characterized by greater airway compromise, milder emphysema, and slower rate of lung function decline than TS-COPD. However, it is unclear if these two etiotypes of COPD have differences in sputum biomarker concentrations. Objective was to compare sputum levels of selected sputum biomarkers between WS-COPD and TS-COPD, and healthy controls. Methods: Eighty-eight women (69±12 years) were recruited and classified into: WS-COPD (n=31), TS-COPD (n=29) and controls (n=28). Using ELISA, we determined induced sputum levels of metalloproteinase 9 (MMP-9), chemokine ligand 5 (CCL5), interleukin-8 (IL-8), chemokine ligand 16 (CCL16/HCC-4) and vascular endothelial growth factor (VEGF-1). Differences were analyzed by Kruskal-Wallis and Mann-Whitney-U tests and correlation between airflow limitation and biomarkers by Spearman's test. Results: At similar degree of airflow obstruction, anthropometrics and medications use, the level of sputum CCL5 was higher in TS-COPD than WS-COPD (p=0.03) without differences in MMP-9, IL-8, CCL16/HCC-4, and VEGF-1. Women with WS-COPD and TS-COPD showed significantly higher sputum levels of MMP-9, IL-8 and CCL5 compared with controls (p<0.001). FEV1% predicted correlated negatively with levels of MMP-9 (rho:-0.26; P=0.016), CCL5 (rho:-0.37; P=0.001), IL-8 (rho:-0.42; P<0.001) and VEGF (rho:-0.22; P=0.04). Conclusion: While sputum concentrations of MMP-9, IL-8, and CCL5 were higher in COPD women compared with controls, women with TS-COPD had higher levels of CCL5 compared with those with WS-COPD. Whether this finding relates to differences in pathobiological pathways remains to be determined.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Enfermedad Pulmonar Obstructiva Crónica , Contaminación por Humo de Tabaco , Humanos , Femenino , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Interleucina-8/metabolismo , Esputo/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Madera , Metaloproteinasa 9 de la Matriz/metabolismo , Carcinoma Hepatocelular/metabolismo , Ligandos , Neoplasias Hepáticas/metabolismo , Humo/efectos adversos , Biomarcadores/metabolismo , Quimiocinas/metabolismo , Productos de Tabaco
2.
Value Health Reg Issues ; 39: 6-13, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37967491

RESUMEN

OBJECTIVES: This study aimed to describe clinical characteristics and direct medical costs associated with disease treatment in Colombia patients with asthma from 1 healthcare provider. METHODS: This was a descriptive study with a retrospective data collection from a healthcare provider's electronic medical records in Colombia. A clinical, demographic, and healthcare resource utilization profile was developed over a 12-month observation period after the identification of eligible patients. To determine the mean cost per patient per year, the total frequencies of resource utilization were added, and the result was multiplied by the unit cost of each of them. RESULTS: A total of 7919 patients were included in the analysis. The mean ± SD cost per patient per year ranged from $189.5 ± $1.900.6 to $240.2 ± $1.903.6 depending on the price guidebook. The total cost had been driven by the medication use (79% of total cost) and by the outpatient visits (20% of total cost). CONCLUSIONS: In the population analyzed, the mean total direct cost per patient per year of asthma was $189.5 and $240.2, depending on the cost source. Direct medical costs were higher in cases classified as severe and in the adult and elderly population. When comparing the sources of resource utilization, it was found that the mean cost per patient obtained from real-life data is lower than the theoretical cost obtained from the bottom-up method with quantification of resources from experts. It is important to consider limitations related to study design and the evolving landscape of asthma treatments.


Asunto(s)
Asma , Adulto , Humanos , Anciano , Colombia , Estudios Retrospectivos , Costos y Análisis de Costo , Asma/tratamiento farmacológico , Atención a la Salud
5.
Physiol Rep ; 6(15): e13797, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30084231

RESUMEN

Low serum CC16 levels are associated with accelerated lung function decline in human population studies, but it is not known whether low serum CC16 levels contribute to lung function decline, or are an epiphenomenon. We tested the hypothesis that unchallenged Cc16-/- mice develop accelerated rates of pulmonary function test abnormalities and pulmonary pathologies over time compared with unchallenged WT mice. Respiratory mechanics, airspace enlargement, and small airway fibrosis were measured in unchallenged wild-type (WT) versus Cc16-/- mice over 6-18 months of age. Lung leukocyte counts and lung levels of metalloproteinases (Mmps), cytokines, oxidative stress, cellular senescence markers (p19 and p21), and lung cell apoptosis, and serum C-reactive protein (CRP) levels were measured in age-matched WT versus Cc16-/- mice. Unchallenged Cc16-/- mice developed greater increases in lung compliance, airspace enlargement, and small airway fibrosis than age-matched WT mice over 6-18 months of age. Cc16-/- mice had greater: (1) lung leukocyte counts; (2) lung levels of Ccl2, Ccl-5, interleukin-10, Mmp-2, and Mmp-9; (3) pulmonary oxidative stress levels, (4) alveolar septal cell apoptosis and staining for p16 and p21; and (5) serum CRP levels. Unchallenged Cc16-/- mice had greater nuclear factor-κB (NF-κB) activation in their lungs than age-matched WT mice, but similar lung levels of secretory phospholipase-A2 activity. Cc16 deficiency in mice leads spontaneously to an accelerated lung aging phenotype with exaggerated pulmonary inflammation and COPD-like lung pathologies associated with increased activation of NF- κB in the lung. CC16 augmentation strategies may reduce lung aging in CC16-deficient individuals.


Asunto(s)
Neumonía/metabolismo , Uteroglobina/fisiología , Resistencia de las Vías Respiratorias/fisiología , Animales , Apoptosis/fisiología , Líquido del Lavado Bronquioalveolar/citología , Citocinas/metabolismo , Femenino , Recuento de Leucocitos , Pulmón/metabolismo , Pulmón/patología , Rendimiento Pulmonar/fisiología , Masculino , Ratones Noqueados , FN-kappa B/metabolismo , Estrés Oxidativo/fisiología , Neumonía/patología , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Mecánica Respiratoria/fisiología , Uteroglobina/deficiencia
6.
Respir Care ; 62(8): 1058-1066, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28559464

RESUMEN

BACKGROUND: The evidence indicates that risk factors other than smoking are important in the development of COPD. It has been postulated that less traditional risk factors (eg, exposure to coal and/or biomass smoke) may interact with smoking to further increase COPD risk. This analysis evaluated the effect of exposure to biomass and smoking on COPD risk in a primary care setting in Latin America. METHODS: Subjects attending routine primary care visits, ≥40 y old, who were current or former smokers or were exposed to biomass smoke, completed a questionnaire and performed spirometry. COPD was defined as post-bronchodilator FEV1/FVC < 0.70 and the lower limit of normal. Smoking was defined by pack-years (≤ 20, 20-30, or > 30), and biomass exposure was defined as an exposure to coal or wood (for heating, cooking, or both) for ≥ 10 y. RESULTS: One thousand seven hundred forty-three individuals completed the questionnaire, and 1,540 performed spirometry. Irrespective of COPD definition, approximately 40% of COPD subjects reported exposure to biomass versus 30% of those without COPD. A higher proportion of COPD subjects (post-bronchodilator FEV1/FVC < 0.70) than those without COPD smoked > 30 pack-years (66% vs 39%); similar results were found with the lower limit of normal definition. Analysis of exposure to biomass > 10 y plus smoking > 20 pack-years (reference was no exposure) found that tobacco smoking (crude odds ratio [OR] 4.50, 95% CI 2.73-7.41; adjusted OR 3.30, 95% CI 1.93-5.63) and biomass exposure (crude OR 3.66, 95% CI 2.00-6.73; adjusted OR 2.28, 95% CI 1.18-4.41) were risk factors for COPD, with smoking a possible confounder for the association between biomass and COPD (post-bronchodilator FEV1/FVC < 0.70); similar results were found with the lower limit of normal definition. CONCLUSIONS: Subjects with COPD from primary care had a higher exposure to biomass and smoking compared with non-COPD subjects. Smoking and biomass are both risk factors for COPD, but they do not appear to have an additive effect.


Asunto(s)
Biomasa , Exposición a Riesgos Ambientales/efectos adversos , Enfermedad Pulmonar Obstructiva Crónica/etiología , Humo/efectos adversos , Fumar/efectos adversos , Adulto , Carbón Mineral , Femenino , Volumen Espiratorio Forzado , Humanos , América Latina , Masculino , Persona de Mediana Edad , Atención Primaria de Salud , Factores de Riesgo , Espirometría , Capacidad Vital , Madera
7.
BMC Pulm Med ; 17(1): 69, 2017 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-28431499

RESUMEN

BACKGROUND: Asthma-COPD overlap syndrome (ACOS) prevalence varies depending on the studied population and definition criteria. The prevalence and clinical characteristics of ACOS in an at-risk COPD primary care population from Latin America was assessed. METHODS: Patients ≥40 years, current/ex-smokers and/or exposed to biomass, attending routine primary care visits completed a questionnaire and performed spirometry. COPD was defined as post-bronchodilator forced expiratory volume in 1 s/forced vital capacity (FEV1/FVC) < 0.70; asthma was defined as either prior asthma diagnosis or wheezing in the last 12 months plus reversibility (increase in post-bronchodilator FEV1 or FVC ≥200 mL and ≥12%); ACOS was defined using a combination of COPD with the two asthma definitions. Exacerbations in the past year among the subgroups were evaluated. RESULTS: One thousand seven hundred forty three individuals completed the questionnaire, 1540 performed acceptable spirometry, 309 had COPD, 231 had prior asthma diagnosis, and 78 asthma by wheezing + reversibility. ACOS prevalence in the total population (by post-bronchodilator FEV1/FVC < 0.70 plus asthma diagnosis) was 5.3 and 2.3% by post-bronchodilator FEV1/FVC < 0.70 plus wheezing + reversibility. In the obstructive population (asthma or COPD), prevalence rises to 17.9 and 9.9% by each definition, and to 26.5 and 11.3% in the COPD population. ACOS patients defined by post-bronchodilator FEV1/FVC < 0.7 plus wheezing + reversibility had the lowest lung function measurements. Exacerbations for ACOS showed a prevalence ratio of 2.68 and 2.20 (crude and adjusted, p < 0.05, respectively) (reference COPD). CONCLUSIONS: ACOS prevalence in primary care varied according to definition used. ACOS by post-bronchodilator FEV1/FVC < 0.7 plus wheezing + reversibility represents a clinical phenotype with more frequent exacerbations, which is probably associated with a different management approach.


Asunto(s)
Asma/epidemiología , Enfermedad Pulmonar Obstructiva Crónica/epidemiología , Adulto , Animales , Asma/tratamiento farmacológico , Broncodilatadores/uso terapéutico , Femenino , Volumen Espiratorio Forzado , Humanos , América Latina/epidemiología , Masculino , Persona de Mediana Edad , Prevalencia , Atención Primaria de Salud , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Análisis de Regresión , Ruidos Respiratorios/fisiopatología , Factores de Riesgo , Espirometría , Encuestas y Cuestionarios , Capacidad Vital
8.
Am J Respir Crit Care Med ; 195(11): 1464-1476, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28085500

RESUMEN

RATIONALE: Patients with chronic obstructive pulmonary disease (COPD) frequently have albuminuria (indicative of renal endothelial cell injury) associated with hypoxemia. OBJECTIVES: To determine whether (1) cigarette smoke (CS)-induced pulmonary and renal endothelial cell injury explains the association between albuminuria and COPD, (2) CS-induced albuminuria is linked to increases in the oxidative stress-advanced glycation end products (AGEs) receptor for AGEs (RAGE) pathway, and (3) enalapril (which has antioxidant properties) limits the progression of pulmonary and renal injury by reducing activation of the AGEs-RAGE pathway in endothelial cells in both organs. METHODS: In 26 patients with COPD, 24 ever-smokers without COPD, 32 nonsmokers who underwent a renal biopsy or nephrectomy, and in CS-exposed mice, we assessed pathologic and ultrastructural renal lesions, and measured urinary albumin/creatinine ratios, tissue oxidative stress levels, and AGEs and RAGE levels in pulmonary and renal endothelial cells. The efficacy of enalapril on pulmonary and renal lesions was assessed in CS-exposed mice. MEASUREMENTS AND MAIN RESULTS: Patients with COPD and/or CS-exposed mice had chronic renal injury, increased urinary albumin/creatinine ratios, and increased tissue oxidative stress and AGEs-RAGE levels in pulmonary and renal endothelial cells. Treating mice with enalapril attenuated CS-induced increases in urinary albumin/creatinine ratios, tissue oxidative stress levels, endothelial cell AGEs and RAGE levels, pulmonary and renal cell apoptosis, and the progression of chronic renal and pulmonary lesions. CONCLUSIONS: Patients with COPD and/or CS-exposed mice have pulmonary and renal endothelial cell injury linked to increased endothelial cell AGEs and RAGE levels. Albuminuria could identify patients with COPD in whom angiotensin-converting enzyme inhibitor therapy improves renal and lung function by reducing endothelial injury.


Asunto(s)
Endotelio/fisiopatología , Riñón/fisiopatología , Pulmón/fisiopatología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Anciano , Animales , Modelos Animales de Enfermedad , Endotelio/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Estrés Oxidativo , Proyectos Piloto
9.
Int J Chron Obstruct Pulmon Dis ; 11: 3059-3067, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27994446

RESUMEN

BACKGROUND: COPD, asthma, and asthma-COPD overlap increase health care resource consumption, predominantly because of hospitalization for exacerbations and also increased visits to general practitioners (GPs) or specialists. Little information is available regarding this in the primary care setting. OBJECTIVES: To describe the prevalence and number of GP and specialist visits for any cause or due to exacerbations in patients with COPD, asthma, and asthma-COPD overlap. METHODS: COPD was defined as post-bronchodilator forced expiratory volume in 1 second/forced vital capacity (FEV1/FVC) ratio <0.70; asthma was defined as prior medical diagnosis, wheezing in the last 12 months, or wheezing plus reversibility (post-bronchodilator FEV1 or FVC increase ≥200 mL and ≥12%); asthma-COPD overlap was defined as post-bronchodilator FEV1/FVC <0.70 plus prior asthma diagnosis. Health care utilization was evaluated as GP and/or specialist visits in the previous year. RESULTS: Among the 1,743 individuals who completed the questionnaire, 1,540 performed acceptable spirometry. COPD patients had a higher prevalence of any medical visits to any physician versus those without COPD (37.2% vs 21.8%, respectively) and exacerbations doubled the number of visits. The prevalence of any medical visits to any physician was also higher in asthma patients versus those without asthma (wheezing: 47.2% vs 22.7%; medical diagnosis: 54.6% vs 21.6%; wheezing plus reversibility: 46.2% vs 23.8%, respectively). Asthma patients with exacerbations had twice the number of visits versus those without an exacerbation. The number of visits was higher (2.8 times) in asthma-COPD overlap, asthma (1.9 times), or COPD (1.4 times) patients versus those without these respiratory diseases; the number of visits due to exacerbation was also higher (4.9 times) in asthma-COPD overlap, asthma (3.5 times), and COPD (3.8 times) patients. CONCLUSION: COPD, asthma, and asthma-COPD overlap increase the prevalence of medical visits and, therefore, health care resource utilization. Attempts to reduce health care resource use in these patients require interventions aimed at preventing exacerbations.


Asunto(s)
Obstrucción de las Vías Aéreas/terapia , Asma/terapia , Recursos en Salud/estadística & datos numéricos , Pulmón/fisiopatología , Atención Primaria de Salud/estadística & datos numéricos , Enfermedad Pulmonar Obstructiva Crónica/terapia , Adulto , Obstrucción de las Vías Aéreas/diagnóstico , Obstrucción de las Vías Aéreas/fisiopatología , Asma/diagnóstico , Asma/fisiopatología , Estudios Transversales , Progresión de la Enfermedad , Femenino , Volumen Espiratorio Forzado , Medicina General , Humanos , Masculino , Persona de Mediana Edad , Visita a Consultorio Médico/estadística & datos numéricos , Fenotipo , Valor Predictivo de las Pruebas , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Derivación y Consulta/estadística & datos numéricos , Factores de Riesgo , Índice de Severidad de la Enfermedad , América del Sur , Espirometría/estadística & datos numéricos , Encuestas y Cuestionarios/estadística & datos numéricos , Síndrome , Factores de Tiempo , Resultado del Tratamiento , Capacidad Vital
10.
Proc Natl Acad Sci U S A ; 113(32): E4681-7, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27444019

RESUMEN

Genetic variants in Hedgehog interacting protein (HHIP) have consistently been associated with the susceptibility to develop chronic obstructive pulmonary disease and pulmonary function levels, including the forced expiratory volume in 1 s (FEV1), in general population samples by genome-wide association studies. However, in vivo evidence connecting Hhip to age-related FEV1 decline and emphysema development is lacking. Herein, using Hhip heterozygous mice (Hhip(+/-)), we observed increased lung compliance and spontaneous emphysema in Hhip(+/-) mice starting at 10 mo of age. This increase was preceded by increases in oxidative stress levels in the lungs of Hhip(+/-) vs. Hhip(+/+) mice. To our knowledge, these results provide the first line of evidence that HHIP is involved in maintaining normal lung function and alveolar structures. Interestingly, antioxidant N-acetyl cysteine treatment in mice starting at age of 5 mo improved lung function and prevented emphysema development in Hhip(+/-) mice, suggesting that N-acetyl cysteine treatment limits the progression of age-related emphysema in Hhip(+/-) mice. Therefore, reduced lung function and age-related spontaneous emphysema development in Hhip(+/-) mice may be caused by increased oxidative stress levels in murine lungs as a result of haploinsufficiency of Hhip.


Asunto(s)
Proteínas Portadoras/genética , Enfisema/etiología , Haploinsuficiencia , Glicoproteínas de Membrana/genética , Acetilcisteína/farmacología , Factores de Edad , Animales , Glutatión/metabolismo , Gutatión-S-Transferasa pi/fisiología , Pulmón/patología , Pulmón/fisiología , Rendimiento Pulmonar , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo
11.
Am J Respir Crit Care Med ; 194(2): 185-97, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-26862784

RESUMEN

RATIONALE: A genetic locus within the FAM13A gene has been consistently associated with chronic obstructive pulmonary disease (COPD) in genome-wide association studies. However, the mechanisms by which FAM13A contributes to COPD susceptibility are unknown. OBJECTIVES: To determine the biologic function of FAM13A in human COPD and murine COPD models and discover the molecular mechanism by which FAM13A influences COPD susceptibility. METHODS: Fam13a null mice (Fam13a(-/-)) were generated and exposed to cigarette smoke. The lung inflammatory response and airspace size were assessed in Fam13a(-/-) and Fam13a(+/+) littermate control mice. Cellular localization of FAM13A protein and mRNA levels of FAM13A in COPD lungs were assessed using immunofluorescence, Western blotting, and reverse transcriptase-polymerase chain reaction, respectively. Immunoprecipitation followed by mass spectrometry identified cellular proteins that interact with FAM13A to reveal insights on FAM13A's function. MEASUREMENTS AND MAIN RESULTS: In murine and human lungs, FAM13A is expressed in airway and alveolar type II epithelial cells and macrophages. Fam13a null mice (Fam13a(-/-)) were resistant to chronic cigarette smoke-induced emphysema compared with Fam13a(+/+) mice. In vitro, FAM13A interacts with protein phosphatase 2A and recruits protein phosphatase 2A with glycogen synthase kinase 3ß and ß-catenin, inducing ß-catenin degradation. Fam13a(-/-) mice were also resistant to elastase-induced emphysema, and this resistance was reversed by coadministration of a ß-catenin inhibitor, suggesting that FAM13A could increase the susceptibility of mice to emphysema development by inhibiting ß-catenin signaling. Moreover, human COPD lungs had decreased protein levels of ß-catenin and increased protein levels of FAM13A. CONCLUSIONS: We show that FAM13A may influence COPD susceptibility by promoting ß-catenin degradation.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , beta Catenina/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estabilidad Proteica , Transducción de Señal , beta Catenina/genética , beta Catenina/fisiología
12.
Expert Opin Ther Targets ; 20(7): 869-83, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26781659

RESUMEN

INTRODUCTION: Club cell protein 16 (CC16) is the most abundant protein in bronchoalveolar lavage fluid. CC16 has anti-inflammatory properties in smoke-exposed lungs, and chronic obstructive pulmonary disease (COPD) is associated with CC16 deficiency. Herein, we explored whether CC16 is a therapeutic target for COPD. AREAS COVERED: We reviewed the literature on the factors that regulate airway CC16 expression, its biologic functions and its protective activities in smoke-exposed lungs using PUBMED searches. We generated hypotheses on the mechanisms by which CC16 limits COPD development, and discuss its potential as a new therapeutic approach for COPD. EXPERT OPINION: CC16 plasma and lung levels are reduced in smokers without airflow obstruction and COPD patients. In COPD patients, airway CC16 expression is inversely correlated with severity of airflow obstruction. CC16 deficiency increases smoke-induced lung pathologies in mice by its effects on epithelial cells, leukocytes, and fibroblasts. Experimental augmentation of CC16 levels using recombinant CC16 in cell culture systems, plasmid and adenoviral-mediated over-expression of CC16 in epithelial cells or smoke-exposed murine airways reduces inflammation and cellular injury. Additional studies are necessary to assess the efficacy of therapies aimed at restoring airway CC16 levels as a new disease-modifying therapy for COPD patients.


Asunto(s)
Terapia Molecular Dirigida , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Uteroglobina/administración & dosificación , Animales , Antiinflamatorios/farmacología , Líquido del Lavado Bronquioalveolar , Diseño de Fármacos , Células Epiteliales/metabolismo , Humanos , Ratones , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Índice de Severidad de la Enfermedad , Fumar/efectos adversos , Uteroglobina/metabolismo
13.
Nat Med ; 22(2): 163-74, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26752519

RESUMEN

Chronic obstructive pulmonary disease (COPD) is linked to both cigarette smoking and genetic determinants. We have previously identified iron-responsive element-binding protein 2 (IRP2) as an important COPD susceptibility gene and have shown that IRP2 protein is increased in the lungs of individuals with COPD. Here we demonstrate that mice deficient in Irp2 were protected from cigarette smoke (CS)-induced experimental COPD. By integrating RNA immunoprecipitation followed by sequencing (RIP-seq), RNA sequencing (RNA-seq), and gene expression and functional enrichment clustering analysis, we identified Irp2 as a regulator of mitochondrial function in the lungs of mice. Irp2 increased mitochondrial iron loading and levels of cytochrome c oxidase (COX), which led to mitochondrial dysfunction and subsequent experimental COPD. Frataxin-deficient mice, which had higher mitochondrial iron loading, showed impaired airway mucociliary clearance (MCC) and higher pulmonary inflammation at baseline, whereas mice deficient in the synthesis of cytochrome c oxidase, which have reduced COX, were protected from CS-induced pulmonary inflammation and impairment of MCC. Mice treated with a mitochondrial iron chelator or mice fed a low-iron diet were protected from CS-induced COPD. Mitochondrial iron chelation also alleviated CS-induced impairment of MCC, CS-induced pulmonary inflammation and CS-associated lung injury in mice with established COPD, suggesting a critical functional role and potential therapeutic intervention for the mitochondrial-iron axis in COPD.


Asunto(s)
Bronquitis/genética , Quelantes del Hierro/farmacología , Proteínas de Unión a Hierro/genética , Hierro/metabolismo , Pulmón/metabolismo , Mitocondrias/metabolismo , Nicotiana , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfisema Pulmonar/genética , Humo/efectos adversos , Anciano , Anciano de 80 o más Años , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Bronquitis/etiología , Modelos Animales de Enfermedad , Complejo IV de Transporte de Electrones/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Proteína 2 Reguladora de Hierro/genética , Proteína 2 Reguladora de Hierro/metabolismo , Hierro de la Dieta , Pulmón/efectos de los fármacos , Lesión Pulmonar/etiología , Lesión Pulmonar/genética , Potencial de la Membrana Mitocondrial , Ratones , Ratones Noqueados , Microscopía Confocal , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Mitocondrias/efectos de los fármacos , Depuración Mucociliar/genética , Neumonía/etiología , Neumonía/genética , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfisema Pulmonar/etiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Fumar/efectos adversos , Frataxina
14.
J Vis Exp ; (95): 52236, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25651034

RESUMEN

COPD is projected to be the third most common cause of mortality world-wide by 2020((1)). Animal models of COPD are used to identify molecules that contribute to the disease process and to test the efficacy of novel therapies for COPD. Researchers use a number of models of COPD employing different species including rodents, guinea-pigs, rabbits, and dogs((2)). However, the most widely-used model is that in which mice are exposed to cigarette smoke. Mice are an especially useful species in which to model COPD because their genome can readily be manipulated to generate animals that are either deficient in, or over-express individual proteins. Studies of gene-targeted mice that have been exposed to cigarette smoke have provided valuable information about the contributions of individual molecules to different lung pathologies in COPD((3-5)). Most studies have focused on pathways involved in emphysema development which contributes to the airflow obstruction that is characteristic of COPD. However, small airway fibrosis also contributes significantly to airflow obstruction in human COPD patients((6)), but much less is known about the pathogenesis of this lesion in smoke-exposed animals. To address this knowledge gap, this protocol quantifies both emphysema development and small airway fibrosis in smoke-exposed mice. This protocol exposes mice to CS using a whole-body exposure technique, then measures respiratory mechanics in the mice, inflates the lungs of mice to a standard pressure, and fixes the lungs in formalin. The researcher then stains the lung sections with either Gill's stain to measure the mean alveolar chord length (as a readout of emphysema severity) or Masson's trichrome stain to measure deposition of extracellular matrix (ECM) proteins around small airways (as a readout of small airway fibrosis). Studies of the effects of molecular pathways on both of these lung pathologies will lead to a better understanding of the pathogenesis of COPD.


Asunto(s)
Alveolos Pulmonares/patología , Enfisema Pulmonar/etiología , Enfisema Pulmonar/patología , Fumar/patología , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Pulmón/patología , Ratones , Enfermedad Pulmonar Obstructiva Crónica/patología
15.
Eur Respir J ; 45(6): 1544-56, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25700379

RESUMEN

Club cell secretory protein-16 (CC16) is the major secreted product of airway club cells, but its role in the pathogenesis of chronic obstructive pulmonary disease (COPD) is unclear. We measured CC16 airway expression in humans with and without COPD and CC16 function in a cigarette smoke (CS)-induced COPD murine model. Airway CC16 expression was measured in COPD patients, smokers without COPD and non-smokers. We exposed wildtype (WT) and CC16(-/-)mice to CS or air for up to 6 months, and measured airway CC16 expression, pulmonary inflammation, alveolar septal cell apoptosis, airspace enlargement, airway mucin 5AC (MUC5AC) expression, small airway remodelling and pulmonary function. Smokers and COPD patients had reduced airway CC16 immunostaining that decreased with increasing COPD severity. Exposing mice to CS reduced airway CC16 expression. CC16(-/-) mice had greater CS-induced emphysema, airway remodelling, pulmonary inflammation, alveolar cell apoptosis, airway MUC5AC expression, and more compliant lungs than WT mice. These changes were associated with increased nuclear factor-κB (NF-κB) activation in CC16(-/-) lungs. CS-induced acute pulmonary changes were reversed by adenoviral-mediated over-expression of CC16. CC16 protects lungs from CS-induced injury by reducing lung NF-κB activation. CS-induced airway CC16 deficiency increases CS-induced pulmonary inflammation and injury and likely contributes to the pathogenesis of COPD.


Asunto(s)
Pulmón/metabolismo , FN-kappa B/metabolismo , Nicotiana , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Humo , Fumar/metabolismo , Uteroglobina/genética , Uteroglobina/metabolismo , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Apoptosis , Estudios de Casos y Controles , Caspasa 3/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Técnicas de Sustitución del Gen , Humanos , Pulmón/fisiopatología , Ratones , Ratones Noqueados , Mucina 5AC/metabolismo , Estrés Oxidativo , Fosfolipasas A2 Secretoras/metabolismo , Alveolos Pulmonares/citología , Enfisema Pulmonar/metabolismo , Pruebas de Función Respiratoria , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
16.
J Immunol ; 193(5): 2469-82, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25063875

RESUMEN

A disintegrin and a metalloproteinase domain (ADAM) 9 is known to be expressed by monocytes and macrophages. In this study, we report that ADAM9 is also a product of human and murine polymorphonuclear neutrophils (PMNs). ADAM9 is not synthesized de novo by circulating PMNs. Rather, ADAM9 protein is stored in the gelatinase and specific granules and the secretory vesicles of human PMNs. Unstimulated PMNs express minimal quantities of surface ADAM9, but activation of PMNs with degranulating agonists rapidly (within 15 min) increases PMN surface ADAM9 levels. Human PMNs produce small quantities of soluble forms of ADAM9. Surprisingly, ADAM9 degrades several extracellular matrix (ECM) proteins, including fibronectin, entactin, laminin, and insoluble elastin, as potently as matrix metalloproteinase-9. However, ADAM9 does not degrade types I, III, or IV collagen or denatured collagens in vitro. To determine whether Adam9 regulates PMN recruitment or ECM protein turnover during inflammatory responses, we compared wild-type and Adam9(-/-) mice in bacterial LPS- and bleomycin-mediated acute lung injury (ALI). Adam9 lung levels increase 10-fold during LPS-mediated ALI in wild-type mice (due to increases in leukocyte-derived Adam9), but Adam9 does not regulate lung PMN (or macrophage) counts during ALI. Adam9 increases mortality, promotes lung injury, reduces lung compliance, and increases degradation of lung elastin during LPS- and/or bleomycin-mediated ALI. Adam9 does not regulate collagen accumulation in the bleomycin-treated lung. Thus, ADAM9 is expressed in an inducible fashion on PMN surfaces where it degrades some ECM proteins, and it promotes alveolar-capillary barrier injury during ALI in mice.


Asunto(s)
Proteínas ADAM/inmunología , Lesión Pulmonar Aguda/inmunología , Matriz Extracelular/inmunología , Proteínas de la Membrana/inmunología , Neutrófilos/inmunología , Proteolisis , Proteínas ADAM/genética , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Animales , Antibióticos Antineoplásicos/efectos adversos , Antibióticos Antineoplásicos/farmacología , Bleomicina/efectos adversos , Bleomicina/farmacología , Barrera Alveolocapilar/inmunología , Barrera Alveolocapilar/patología , Colágeno/genética , Colágeno/inmunología , Elastina/genética , Elastina/inmunología , Matriz Extracelular/genética , Humanos , Lipopolisacáridos/toxicidad , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Neutrófilos/patología
17.
PLoS One ; 9(5): e97485, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24828408

RESUMEN

OBJECTIVES: Matrix metalloproteinase-8 (MMP-8) promotes lung fibrotic responses to bleomycin in mice. Although prior studies reported that MMP-8 levels are increased in plasma and bronchoalveolar lavage fluid (BALF) samples from IPF patients, neither the bioactive forms nor the cellular sources of MMP-8 in idiopathic pulmonary fibrosis (IPF) patients have been identified. It is not known whether MMP-8 expression is dys-regulated in IPF leukocytes or whether MMP-8 plasma levels correlate with IPF outcomes. Our goal was to address these knowledge gaps. METHODS: We measured MMP-8 levels and forms in blood and lung samples from IPF patients versus controls using ELISAs, western blotting, and qPCR, and assessed whether MMP-8 plasma levels in 73 IPF patients correlate with rate of lung function decline and mortality. We used immunostaining to localize MMP-8 expression in IPF lungs. We quantified MMP-8 levels and forms in blood leukocytes from IPF patients versus controls. RESULTS: IPF patients have increased BALF, whole lung, and plasma levels of soluble MMP-8 protein. Active MMP-8 is the main form elevated in IPF lungs. MMP-8 mRNA levels are increased in monocytes from IPF patients, but IPF patients and controls have similar levels of MMP-8 in PMNs. Surprisingly, macrophages and airway epithelial cells are the main cells expressing MMP-8 in IPF lungs. Plasma and BALF MMP-8 levels do not correlate with decline in lung function and/or mortality in IPF patients. CONCLUSION: Blood and lung MMP-8 levels are increased in IPF patients. Active MMP-8 is the main form elevated in IPF lungs. Surprisingly, blood monocytes, lung macrophages, and airway epithelial cells are the main cells in which MMP-8 is upregulated in IPF patients. Plasma and BALF MMP-8 levels are unlikely to serve as a prognostic biomarker for IPF patients. These results provide new information about the expression patterns of MMP-8 in IPF patients.


Asunto(s)
Células Epiteliales/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/metabolismo , Metaloproteinasa 8 de la Matriz/metabolismo , Sistema Mononuclear Fagocítico/metabolismo , Anciano , Líquido del Lavado Bronquioalveolar/química , Femenino , Humanos , Leucocitos/metabolismo , Macrófagos Alveolares/metabolismo , Masculino , Metaloproteinasa 8 de la Matriz/sangre , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA