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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Toxicol Environ Health B Crit Rev ; 26(5): 275-305, 2023 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-37183431

RESUMEN

Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide, and its global health burden is increasing. COPD is characterized by emphysema, mucus hypersecretion, and persistent lung inflammation, and clinically by chronic airflow obstruction and symptoms of dyspnea, cough, and fatigue in patients. A cluster of pathologies including chronic bronchitis, emphysema, asthma, and cardiovascular disease in the form of hypertension and atherosclerosis variably coexist in COPD patients. Underlying causes for COPD include primarily tobacco use but may also be driven by exposure to air pollutants, biomass burning, and workplace related fumes and chemicals. While no single animal model might mimic all features of human COPD, a wide variety of published models have collectively helped to improve our understanding of disease processes involved in the genesis and persistence of COPD. In this review, the pathogenesis and associated risk factors of COPD are examined in different mammalian models of the disease. Each animal model included in this review is exclusively created by tobacco smoke (TS) exposure. As animal models continue to aid in defining the pathobiological mechanisms of and possible novel therapeutic interventions for COPD, the advantages and disadvantages of each animal model are discussed.


Asunto(s)
Enfisema , Enfermedad Pulmonar Obstructiva Crónica , Enfisema Pulmonar , Contaminación por Humo de Tabaco , Animales , Humanos , Contaminación por Humo de Tabaco/efectos adversos , Enfermedad Pulmonar Obstructiva Crónica/epidemiología , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/patología , Humo/efectos adversos , Enfisema Pulmonar/inducido químicamente , Enfisema Pulmonar/complicaciones , Enfisema/inducido químicamente , Enfisema/complicaciones , Modelos Animales de Enfermedad , Mamíferos
2.
Allergy ; 77(4): 1139-1149, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34525218

RESUMEN

BACKGROUND: Genetic association studies have demonstrated that the SNP rs12603332 located on chromosome 17q21 is highly associated with the risk of the development of asthma. METHODS: To determine whether SNP rs1260332 is functional in regulating levels of ORMDL3 expression, we used a Cytosine Base Editor (CBE) plasmid DNA or a CBE mRNA to edit the rs12603332 C risk allele to the T non-risk allele in a human lymphocyte cell line (i.e., Jurkat cells) and in primary human CD4 T cells that carry the C risk alleles. RESULTS: Jurkat cells with the rs12603332 C risk allele expressed significantly higher levels of ORMDL3 mRNA, as well as the ORMDL3 regulated gene ATF6α as assessed by qPCR compared to Jurkat clones with the T non-risk allele. In primary human CD4 T cells, we edited 90 ± 3% of the rs12603332-C risk allele to the T non-risk allele and observed a reduction in ORMDL3 and ATF6α expression. Bioinformatic analysis predicted that the non-risk allele rs12603332-T could be the central element of the E-box binding motif (CANNTG) recognized by the E47 transcription factor. An EMSA assay confirmed the bioinformatics prediction demonstrating that a rs12603332-T containing probe bound to the transcription factor E47 in vitro. CONCLUSIONS: SNP rs12603332 is functional in regulating the expression of ORMDL3 as well as ORMDL3 regulated gene ATF6α expression. In addition, we demonstrate the use of CBE technology in functionally interrogating asthma-associated SNPs using studies of primary human CD4 cells.


Asunto(s)
Asma , Cromosomas Humanos Par 17 , Citosina , Asma/genética , Estudios de Casos y Controles , Cromosomas , Predisposición Genética a la Enfermedad , Humanos , Proteínas de la Membrana/genética , Polimorfismo de Nucleótido Simple , ARN Mensajero , Factores de Transcripción/genética
3.
Inhal Toxicol ; 32(8): 328-341, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32781858

RESUMEN

OBJECTIVE: Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide and has been associated with periods of intense lung inflammation. The objective of this study was to characterize whether similar rat strains, possessing different genetic predispositions, might play a role in exacerbating the pathophysiology of COPD-like cellular and structural changes with progressive 12-week exposure to tobacco smoke (TS). Normotensive Wistar Kyoto (WKY) and spontaneously hypertensive (SH) rats were compared. MATERIALS AND METHODS: WKY and SH rats were exposed to filtered air or to tobacco smoke at a particulate concentration of 80 mg/m3 for 4, 8, or 12 weeks. Necropsy was performed 24 h after the last exposure to obtain cells by bronchoalveolar lavage for total cell and differential counts. Scoring of lung tissues and immunohistochemical staining for M1 (pro-inflammatory) and M2 (anti-inflammatory) macrophages were performed on paraffin-embedded lung sections. RESULTS AND DISCUSSION: With progressive exposure, TS-exposed SH rats demonstrated significant airspace enlargement, mucin production, and lung inflammation compared to their FA control and TS-matched WKY rats. Moreover, SH rats also demonstrated increased expression of the M1 marker in alveolar macrophages compared to FA control, as well as the M2 marker compared to controls and TS-exposed WKY rats. CONCLUSION: The progressive tobacco smoke exposure contributes to persistent lung injury and inflammation that can be significantly enhanced by rat strain susceptibility in the genesis of COPD.


Asunto(s)
Bronquiolitis/inmunología , Lesión Pulmonar/inmunología , Pulmón/inmunología , Nicotiana , Humo/efectos adversos , Contaminación por Humo de Tabaco/efectos adversos , Animales , Bronquiolitis/patología , Quimiocina CCL2/inmunología , Quimiocina CXCL1/inmunología , Inflamación/inmunología , Inflamación/patología , Pulmón/patología , Lesión Pulmonar/patología , Macrófagos/inmunología , Masculino , Ratas Endogámicas SHR , Ratas Endogámicas WKY
5.
J Immunol ; 195(8): 3546-56, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26355153

RESUMEN

Chronic asthma is associated with airway remodeling and decline in lung function. In this article, we show that follistatin-like 1 (Fstl1), a mediator not previously associated with asthma, is highly expressed by macrophages in the lungs of humans with severe asthma. Chronic allergen-challenged Lys-Cre(tg) /Fstl1(Δ/Δ) mice in whom Fstl1 is inactivated in macrophages/myeloid cells had significantly reduced airway remodeling and reduced levels of oncostatin M (OSM), a cytokine previously not known to be regulated by Fstl1. The importance of the Fstl1 induction of OSM to airway remodeling was demonstrated in murine studies in which administration of Fstl1 induced airway remodeling and increased OSM, whereas administration of an anti-OSM Ab blocked the effect of Fstl1 on inducing airway remodeling, eosinophilic airway inflammation, and airway hyperresponsiveness, all cardinal features of asthma. Overall, these studies demonstrate that the Fstl1/OSM pathway may be a novel pathway to inhibit airway remodeling in severe human asthma.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/inmunología , Asma/inmunología , Proteínas Relacionadas con la Folistatina/inmunología , Oncostatina M/inmunología , Transducción de Señal/inmunología , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Remodelación de las Vías Aéreas (Respiratorias)/genética , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Asma/genética , Asma/patología , Femenino , Proteínas Relacionadas con la Folistatina/genética , Humanos , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones , Oncostatina M/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
6.
Inhal Toxicol ; 28(14): 677-685, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27829308

RESUMEN

CONTEXT: Approximately four million people die every year from chronic obstructive pulmonary disease (COPD), with more than 80% of the cases attributed to smoking. OBJECT: The purpose of this study was to examine the rat strain and sex-related differences and the extended tobacco smoke exposure to induce lung injury and inflammation with the goal of finding a suitable rodent model to study COPD. METHODS: Male and female spontaneously hypertensive (SH) and male Wistar Kyoto (WKY) rats were exposed to filtered air (FA) or to tobacco smoke (TS: 90 mg/m3 particulate concentration) for 6 h/day, three days/week for 4 or 12 weeks. RESULTS: Male SH rats demonstrated an enhanced, persistent inflammatory response compared to female SH and male WKY rats with extended TS exposure. Following four weeks of TS exposure, male SH rats had significantly increased total leukocytes and macrophage numbers, levels of TNF-alpha and elevated lactate dehydrogenase activity in bronchoalveolar lavage fluid compared with female SH, male WKY rats and corresponding controls. After 12 weeks of TS exposure, male SH rats continued to show significant increase in inflammatory cells and TNF-alpha, as well as IL-6 mRNA lung expression. In addition, the alveolar airspace of male SH rats exposed to TS was significantly enlarged compared to their FA controls, female SH and WKY rats. CONCLUSION: The male SH rat demonstrates greater cellular, inflammatory and structural changes highly reminiscent of COPD compared to female SH and male WKY rats, suggesting that the male SH rat is an optimal rodent model to study COPD.


Asunto(s)
Hipertensión , Inflamación/inducido químicamente , Nicotiana/efectos adversos , Humo/efectos adversos , Administración por Inhalación , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Recuento de Células , Citocinas/genética , Citocinas/metabolismo , Femenino , Expresión Génica , Hemo Oxigenasa (Desciclizante)/metabolismo , Hipertensión/genética , Hipertensión/metabolismo , Hipertensión/patología , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Estrés Oxidativo/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Factores Sexuales , Especificidad de la Especie
8.
Proc Natl Acad Sci U S A ; 109(41): 16648-53, 2012 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-23011799

RESUMEN

Orosomucoid like 3 (ORMDL3) has been strongly linked with asthma in genetic association studies, but its function in asthma is unknown. We demonstrate that in mice ORMDL3 is an allergen and cytokine (IL-4 or IL-13) inducible endoplasmic reticulum (ER) gene expressed predominantly in airway epithelial cells. Allergen challenge induces a 127-fold increase in ORMDL3 mRNA in bronchial epithelium in WT mice, with lesser 15-fold increases in ORMDL-2 and no changes in ORMDL-1. Studies of STAT-6-deficient mice demonstrated that ORMDL3 mRNA induction highly depends on STAT-6. Transfection of ORMDL3 in human bronchial epithelial cells in vitro induced expression of metalloproteases (MMP-9, ADAM-8), CC chemokines (CCL-20), CXC chemokines (IL-8, CXCL-10, CXCL-11), oligoadenylate synthetases (OAS) genes, and selectively activated activating transcription factor 6 (ATF6), an unfolded protein response (UPR) pathway transcription factor. siRNA knockdown of ATF-6α in lung epithelial cells inhibited expression of SERCA2b, which has been implicated in airway remodeling in asthma. In addition, transfection of ORMDL3 in lung epithelial cells activated ATF6α and induced SERCA2b. These studies provide evidence of the inducible nature of ORMDL3 ER expression in particular in bronchial epithelial cells and suggest an ER UPR pathway through which ORMDL3 may be linked to asthma.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/metabolismo , Factor de Transcripción Activador 6/metabolismo , Quimiocinas/metabolismo , Pulmón/metabolismo , Proteínas de la Membrana/metabolismo , Metaloproteasas/metabolismo , 2',5'-Oligoadenilato Sintetasa/genética , Factor de Transcripción Activador 6/genética , Animales , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Quimiocinas/genética , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Células Epiteliales/metabolismo , Epitelio/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Interleucina-13/farmacología , Interleucina-4/farmacología , Pulmón/citología , Proteínas de la Membrana/genética , Metaloproteasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovalbúmina/farmacología , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Respuesta de Proteína Desplegada/efectos de los fármacos , Respuesta de Proteína Desplegada/genética
10.
J Immunol ; 188(6): 2622-9, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22327070

RESUMEN

The fungal allergen, Alternaria, is specifically associated with severe asthma, including life-threatening exacerbations. To better understand the acute innate airway response to Alternaria, naive wild-type (WT) mice were challenged once intranasally with Alternaria. Naive WT mice developed significant bronchoalveolar lavage eosinophilia following Alternaria challenge when analyzed 24 h later. In contrast to Alternaria, neither Aspergillus nor Candida induced bronchoalveolar lavage eosinophilia. Gene microarray analysis of airway epithelial cell brushings demonstrated that Alternaria-challenged naive WT mice had a >20-fold increase in the level of expression of found in inflammatory zone 1 (FIZZ1/Retnla), a resistin-like molecule. Lung immunostaining confirmed strong airway epithelial FIZZ1 expression as early as 3 h after a single Alternaria challenge that persisted for ≥5 d and was significantly reduced in STAT6-deficient, but not protease-activated receptor 2-deficient mice. Bone marrow chimera studies revealed that STAT6 expressed in lung cells was required for epithelial FIZZ1 expression, whereas STAT6 present in bone marrow-derived cells contributed to airway eosinophilia. Studies investigating which cells in the nonchallenged lung bind FIZZ1 demonstrated that CD45(+)CD11c(+) cells (macrophages and dendritic cells), as well as collagen-1-producing CD45(-) cells (fibroblasts), can bind to FIZZ1. Importantly, direct administration of recombinant FIZZ1 to naive WT mice led to airway eosinophilia, peribronchial fibrosis, and increased thickness of the airway epithelium. Thus, Alternaria induces STAT6-dependent acute airway eosinophilia and epithelial FIZZ1 expression that promotes airway fibrosis and epithelial thickness. This may provide some insight into the uniquely pathogenic aspects of Alternaria-associated asthma.


Asunto(s)
Alternariosis/patología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Eosinofilia Pulmonar/patología , Fibrosis Pulmonar/patología , Factor de Transcripción STAT6/metabolismo , Alternaria/inmunología , Alternariosis/inmunología , Alternariosis/metabolismo , Animales , Separación Celular , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Péptidos y Proteínas de Señalización Intercelular/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Eosinofilia Pulmonar/inmunología , Eosinofilia Pulmonar/metabolismo , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT6/inmunología
12.
J Immunol ; 182(1): 684-91, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19109202

RESUMEN

We screened bronchoalveolar lavage (BAL) fluids from COPD-E (chronic obstructive pulmonary disease-Emphysema) and control subjects using a 120 Ab cytokine array and demonstrated that adiponectin was highly expressed in BAL in COPD-E. An adiponectin ELISA confirmed that adiponectin was highly expressed in BAL in COPD-E compared with smokers and healthy control subjects. Immunohistochemistry studies of lung sections from subjects with COPD-E demonstrated that airway epithelial cells expressed significant levels of adiponectin and adiponectin receptor (AdipoR) 1 but not AdipoR2. In vitro studies with purified populations of human lung A549 epithelial cells demonstrated that they expressed both adiponectin and AdipoR1 (but not AdipoR2) as assessed by RT-PCR, Western blot, and immunohistochemistry. Lung A549 epithelial AdipoR1were functional as incubation with adiponectin induced release of IL-8, which was inhibited by small interfering RNA to AdipoR1. Using a mouse model of COPD, tobacco smoke exposure induced both evidence of COPD as well as increased levels of adiponectin in BAL fluid and increased adiponectin expression by airway epithelial cells. As adiponectin expression in adipocytes is dependent upon NF-kappaB we determined levels of adiponectin in tobacco smoke exposed CC10-Cre(tg)/Ikkbeta(Delta/Delta) mice (deficient in the ability to activate NF-kappaB in airway epithelium). These studies demonstrated that CC10-Cre(tg)/Ikkbeta(Delta/Delta) and wild-type mice had similar levels of BAL adiponectin and airway epithelial adiponectin immunostaining. Overall, these studies demonstrate the novel observation that adiponectin and functional AdipoR1are expressed by lung epithelial cells, suggesting a potential autocrine and/or paracrine pathway for adiponectin to activate epithelial cells in COPD-E.


Asunto(s)
Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Receptores de Adiponectina/biosíntesis , Mucosa Respiratoria/metabolismo , Adiponectina/biosíntesis , Adiponectina/genética , Adiponectina/fisiología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Interleucina-8/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/fisiología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfisema Pulmonar/inmunología , Enfisema Pulmonar/metabolismo , Enfisema Pulmonar/patología , Receptores de Adiponectina/genética , Receptores de Adiponectina/fisiología , Mucosa Respiratoria/patología , Transducción de Señal/genética , Transducción de Señal/inmunología
13.
J Pediatr Gastroenterol Nutr ; 53(4): 409-16, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21970996

RESUMEN

OBJECTIVES: Eosinophilic esophagitis (EoE) is a disorder characterized histologically by tissue eosinophilia. Sialic acid-binding immunoglobulin-like lectin (Siglec-F) is a receptor highly expressed on mouse eosinophils and mediates eosinophilic apoptosis. We investigated whether administration of an anti-Siglec-F Ab would reduce esophageal eosinophilic inflammation and remodeling in a mouse model of egg ovalbumin (OVA)-induced EoE. SUBJECTS AND METHODS: Three groups of mice were studied (no OVA, OVA + anti-Siglec-F Ab, and OVA + isotype control Ab). Mice were sensitized intraperitoneally and then challenged chronically with intraesophageal OVA. Levels of esophageal eosinophils and features of remodeling (angiogenesis, vascular endothelial growth factor expression, deposition of fibronectin, basal zone hyperplasia, and fibrosis) were quantitated by immunohistochemistry and image analysis. RESULTS: Administration of an anti-Siglec-F Ab to OVA-challenged mice significantly reduced levels of esophageal eosinophils, down to levels noted in non-OVA-challenged mice. The anti-Siglec-F Ab also reduced features of OVA-induced remodeling, including angiogenesis, basal zone hyperplasia, and fibronectin deposition. The reduced angiogenesis in anti-Siglec-F Ab-treated mice was associated with reduced numbers of vascular endothelial growth factor-positive cells in the esophagus. The anti-Siglec-F antibody did not significantly reduce esophageal fibrosis as assessed by trichrome staining. CONCLUSIONS: Administration of an anti-Siglec-F antibody significantly decreased the number of eosinophils in the esophagus in a mouse model of OVA-induced EoE. The reduction in eosinophilic inflammation was associated with a significant decrease in levels of angiogenesis, deposition of fibronectin, and basal zone hyperplasia. Studies in this pre-clinical model of EoE suggest that Siglec-F (and its human paralog Siglec-8) may be novel therapeutic targets to reduce eosinophilic inflammation in EoE.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Anticuerpos/farmacología , Antígenos de Diferenciación Mielomonocítica/metabolismo , Esofagitis Eosinofílica/terapia , Eosinófilos/patología , Animales , Antígenos de Diferenciación Mielomonocítica/inmunología , Apoptosis , Modelos Animales de Enfermedad , Esofagitis Eosinofílica/sangre , Esofagitis Eosinofílica/patología , Eosinófilos/efectos de los fármacos , Femenino , Inflamación/patología , Ratones , Ratones Endogámicos BALB C , Neovascularización Patológica , Ovalbúmina/metabolismo , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo
14.
JCI Insight ; 6(7)2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33661765

RESUMEN

ORM1-like 3 (ORMDL3) has strong genetic linkage to childhood onset asthma. To determine whether ORMDL3 selective expression in airway smooth muscle (ASM) influences ASM function, we used Cre-loxP techniques to generate transgenic mice (hORMDL3Myh11eGFP-cre), which express human ORMDL3 selectively in smooth muscle cells. In vitro studies of ASM cells isolated from the bronchi of hORMDL3Myh11eGFP-cre mice demonstrated that they developed hypertrophy (quantitated by FACS and image analysis), developed hyperplasia (assessed by BrdU incorporation), and expressed increased levels of tropomysin proteins TPM1 and TPM4. siRNA knockdown of TPM1 or TPM4 demonstrated their importance to ORMDL3-mediated ASM proliferation but not hypertrophy. In addition, ASM derived from hORMDL3Myh11eGFP-cre mice had increased contractility to histamine in vitro, which was associated with increased levels of intracellular Ca2+; increased cell surface membrane Orai1 Ca2+ channels, which mediate influx of Ca2+ into the cytoplasm; and increased expression of ASM contractile genes sarco/endoplasmic reticulum Ca2+ ATPase 2b and smooth muscle 22. In vivo studies of hORMDL3Myh11eGFP-cre mice demonstrated that they had a spontaneous increase in ASM and airway hyperreactivity (AHR). ORMDL3 expression in ASM thus induces changes in ASM (hypertrophy, hyperplasia, increased contractility), which may explain the contribution of ORMDL3 to the development of AHR in childhood onset asthma, which is highly linked to ORMDL3 on chromosome 17q12-21.


Asunto(s)
Proteínas de la Membrana/genética , Músculo Liso/patología , Tropomiosina/genética , Animales , Asma/genética , Asma/patología , Hiperreactividad Bronquial/etiología , Calcio/metabolismo , Proliferación Celular , Células Cultivadas , Histamina/farmacología , Humanos , Hiperplasia , Hipertrofia , Ratones Transgénicos , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Músculo Liso/fisiología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Tropomiosina/metabolismo
15.
Am J Physiol Lung Cell Mol Physiol ; 299(6): L834-42, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20935231

RESUMEN

Adiponectin is a cytokine with both proinflammatory and anti-inflammatory properties that is expressed in epithelial cells in the airway in chronic obstructive pulmonary disease-emphysema (COPD-E). To determine whether adiponectin modulates levels of lung inflammation in tobacco smoke-induced COPD-E, we used a mouse model of COPD-E in which either adiponectin-deficient or wild-type (WT) mice were exposed to tobacco smoke for 6 mo. Outcomes associated with tobacco smoke-induced COPD-E were quantitated including lung inflammation [bronchoalveolar lavage (BAL) and total and differential cell count], lung mediators of inflammation (cytokines and chemokines), air space enlargement (i.e., linear intercept), and lung function (tissue elastance) in the different groups of mice. Whereas exposure of WT mice to tobacco smoke for 6 mo induced significant lung inflammation (increased total BAL cells, neutrophils, and macrophages), adiponectin-deficient mice had minimal BAL inflammation when exposed to tobacco smoke for 6 mo. In addition, whereas chronic tobacco-exposed WT mice had significantly increased levels of lung mediators of inflammation [i.e., TNF-α, keratinocyte-derived chemokine (KC), and adiponectin] as well as significantly increased air space enlargement (increased linear intercept) and decreased tissue elastance, exposure of adiponectin-deficient mice to chronic tobacco smoke resulted in no further increase in lung mediators, air space enlargement, or tissue elastance. In vitro studies demonstrated that BAL macrophages derived from adiponectin-deficient mice incubated in media containing tobacco smoke expressed minimal TNF-α or KC compared with BAL macrophages from WT mice. These studies suggest that adiponectin plays an important proinflammatory role in tobacco smoke-induced COPD-E.


Asunto(s)
Nicotiana/efectos adversos , Neumonía , Enfisema Pulmonar , Humo/efectos adversos , Adiponectina/deficiencia , Adiponectina/genética , Adiponectina/farmacología , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Citocinas/inmunología , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Elastasa Pancreática/metabolismo , Neumonía/inducido químicamente , Neumonía/inmunología , Neumonía/patología , Enfisema Pulmonar/inducido químicamente , Enfisema Pulmonar/inmunología , Enfisema Pulmonar/patología
16.
Respir Res ; 11: 154, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21040544

RESUMEN

BACKGROUND: In this study we examined the role of Siglec-F, a receptor highly expressed on eosinophils, in contributing to mucus expression, airway remodeling, and Siglec-F ligand expression utilizing Siglec-F deficient mice exposed to chronic allergen challenge. METHODS: Wild type (WT) and Siglec-F deficient mice were sensitized and challenged chronically with OVA for one month. Levels of airway inflammation (eosinophils), Siglec-F ligand expresion and remodeling (mucus, fibrosis, smooth muscle thickness, extracellular matrix protein deposition) were assessed in lung sections by image analysis and immunohistology. Airway hyperreactivity to methacholine was assessed in intubated and ventilated mice. RESULTS: Siglec-F deficient mice challenged with OVA for one month had significantly increased numbers of BAL and peribronchial eosinophils compared to WT mice which was associated with a significant increase in mucus expression as assessed by the number of periodic acid Schiff positive airway epithelial cells. In addition, OVA challenged Siglec-F deficient mice had significantly increased levels of peribronchial fibrosis (total lung collagen, area of peribronchial trichrome staining), as well as increased numbers of peribronchial TGF-ß1+ cells, and increased levels of expression of the extracellular matrix protein fibronectin compared to OVA challenged WT mice. Lung sections immunostained with a Siglec-Fc to detect Siglec-F ligand expression demonstrated higher levels of expression of the Siglec-F ligand in the peribronchial region in OVA challenged Siglec-F deficient mice compared to WT mice. WT and Siglec-F deficient mice challenged intranasally with IL-4 or IL-13 had significantly increased levels of airway epithelial Siglec-F ligand expression, whereas this was not observed in WT or Siglec-F deficient mice challenged with TNF-α. There was a significant increase in the thickness of the peribronchial smooth muscle layer in OVA challenged Siglec-F deficient mice, but this was not associated with significant increased airway hyperreactivity compared to WT mice. CONCLUSIONS: Overall, this study demonstrates an important role for Siglec-F in modulating levels of chronic eosinophilic airway inflammation, peribronchial fibrosis, thickness of the smooth muscle layer, mucus expression, fibronectin, and levels of peribronchial Siglec-F ligands suggesting that Siglec-F may normally function to limit levels of chronic eosinophilic inflammation and remodeling. In addition, IL-4 and IL-13 are important regulators of Siglec-F ligand expression by airway epithelium.


Asunto(s)
Antígenos de Diferenciación Mielomonocítica/metabolismo , Eosinófilos/metabolismo , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Pulmón/metabolismo , Moco/metabolismo , Ovalbúmina/farmacología , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Animales , Células Cultivadas , Eosinófilos/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Ligandos , Pulmón/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
17.
Toxicol Lett ; 278: 1-8, 2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28698096

RESUMEN

Airborne particulate matter (PM) is associated with adverse cardiorespiratory effects. To better understand source-orientated PM toxicity, a comparative study of the biological effects of fine PM (diameter≤2.5µm, PM2.5) collected during the winter season from Shanxi Province, China, and the Central Valley, California, United States, was conducted. The overarching hypothesis for this study was to test whether the chemical composition of PM on an equal mass basis from two urban areas, one in China and one in California, can lead to significantly different effects of acute toxicity and inflammation in the lungs of healthy young mice. Male, 8-week old BALB/C mice received a single 50µg dose of vehicle, Taiyuan PM or Sacramento PM by oropharyngeal aspiration and were sacrificed 24h later. Bronchoalveolar lavage, ELISA and histopathology were performed along with chemical analysis of PM composition. Sacramento PM had a greater proportion of oxidized organic material, significantly increased neutrophil numbers and elevated CXCL-1 and TNF-α protein levels compared to the Taiyuan PM. The findings suggest that Sacramento PM2.5 was associated with a greater inflammatory response compared to that of Taiyuan PM2.5 that may be due to a higher oxidice. Male, 8-week old BALB/C mice received a single 50µg dose of vehicle, Taiyuan PM or Sacramento PM by oropharyngeal aspiration and were sacrificed 24h later. Bronchoalveolar lavage, ELISA and histopathology were performed along with chemical analysis of PM composition. Sacramento PM had a greater proportion of oxidized organic material, significantly increased neutrophil numbers and elevated CXCL-1 and TNF-α protein levels compared to the Taiyuan PM. The findings suggest that Sacramento PM2.5 was associated with a greater inflammatory response compared to that of Taiyuan PM2.5 that may be due to a higher oxidized state of organic carbon and copper content.


Asunto(s)
Exposición por Inhalación/efectos adversos , Pulmón/efectos de los fármacos , Material Particulado/toxicidad , Neumonía/inducido químicamente , Estaciones del Año , Animales , Líquido del Lavado Bronquioalveolar/inmunología , California , Quimiocina CXCL1/metabolismo , China , Ensayo de Inmunoadsorción Enzimática , Mediadores de Inflamación/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Masculino , Espectrometría de Masas/métodos , Ratones Endogámicos BALB C , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Tamaño de la Partícula , Neumonía/inmunología , Neumonía/metabolismo , Neumonía/patología , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo
18.
Int Immunopharmacol ; 18(1): 35-42, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24239745

RESUMEN

Studies of AMCase inhibition in mouse models of lung eosinophilic inflammation have produced conflicting results with some studies demonstrating inhibition of eosinophilic inflammation and others not. No studies have investigated the role of AMCase inhibition in eosinophilic esophagitis (EoE). We have used a mouse model of egg (OVA) induced EoE to determine whether pharmacologic inhibition of AMCase with allosamidin reduced eosinophilic inflammation and remodeling in the esophagus in EoE. Administration of intra-esophageal OVA for 6weeks to BALB/c mice induced increased levels of esophageal eosinophils, mast cells, and features of esophageal remodeling (fibrosis, basal zone hyperplasia, deposition of the extracellular matrix protein fibronectin). Administration of intraperitoneal (ip) allosamidin to BALB/c mice significantly inhibited AMCase enzymatic activity in the esophagus. Pharmacologic inhibition of AMCase with ip allosamidin inhibited both OVA induced increases in esophageal eosinophilic inflammation and OVA induced esophageal remodeling (fibrosis, epithelial basal zone hyperplasia, extracellular matrix deposition of fibronectin). This inhibition of eosinophilic inflammation in the esophagus by ip allosamidin was associated with reduced eotaxin-1 expression in the esophagus. Oral allosamidin inhibited eosinophilic inflammation in the epithelium but did not inhibit esophageal remodeling. These studies suggest that pharmacologic inhibition of AMCase results in inhibition of eosinophilic inflammation and remodeling in the esophagus in a mouse model of egg induced EoE partially through effects in the esophagus on reducing chemokines (i.e. eotaxin-1) implicated in the pathogenesis of EoE.


Asunto(s)
Acetilglucosamina/análogos & derivados , Quitinasas/metabolismo , Inhibidores Enzimáticos/administración & dosificación , Esofagitis Eosinofílica/tratamiento farmacológico , Esófago/patología , Hipersensibilidad Inmediata/tratamiento farmacológico , Trisacáridos/administración & dosificación , Acetilglucosamina/administración & dosificación , Alérgenos/inmunología , Animales , Células Cultivadas , Quimiocina CCL11/genética , Quimiocina CCL11/metabolismo , Modelos Animales de Enfermedad , Huevos/efectos adversos , Esofagitis Eosinofílica/inducido químicamente , Esofagitis Eosinofílica/inmunología , Esófago/efectos de los fármacos , Esófago/inmunología , Femenino , Fibrosis , Humanos , Hiperplasia , Hipersensibilidad Inmediata/inducido químicamente , Hipersensibilidad Inmediata/inmunología , Mediadores de Inflamación/metabolismo , Ratones Endogámicos BALB C , Terapia Molecular Dirigida , Ovalbúmina/efectos adversos , Ovalbúmina/inmunología
19.
Chest ; 139(6): 1380-1387, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20966041

RESUMEN

BACKGROUND: Tobacco smoking is a principal cause of COPD-emphysema (COPD-E). Whether discontinuing smoking for at least 4 years halts airway inflammation and progression of COPD-E in prior smokers is unknown. In this study we investigated whether discontinuing smoking for approximately 4 years in ex-smokers with GOLD (Global Initiative for Chronic Lung Disease) stage IIb (moderately severe) COPD-E stopped airway inflammation (ie, sputum biomarkers) and halted the progression of COPD-E on chest CT scan. METHODS: Ten ex-smokers with COPD-E who had quit smoking underwent chest CT scans to document the extent of COPD-E, assessment of lung function (FEV(1) and diffusing capacity of lung for carbon monoxide), sputum induction for biomarkers of inflammation (measured by enzyme-linked immunosorbent assay), and blood cotinine levels at baseline and approximately 4 years later. Normal healthy subjects (n = 7) and normal current smokers with no CT scan evidence of COPD-E (n = 8) served as sputum biomarker comparison groups. RESULTS: After approximately 4 years of not smoking (documented by cotinine levels), ex-smokers with COPD-E had persistent increased levels of mediators of inflammation in sputum (myeloperoxidase, leukotriene B4, IL-8, monocyte chemoattractant protein-1, matrix metalloprotease-9), which was associated with significant progression of COPD-E on chest CT scan. CONCLUSIONS: Cessation of tobacco smoking in heavy smokers with moderately severe COPD-E is associated with evidence of persistent airway inflammation and progression of COPD-E on CT scan 4 years later. Discontinuing smoking may slow the rate of progression of moderate severity COPD-E, but it does not prevent persistent airway inflammation and significant progression of COPD-E on CT scan.


Asunto(s)
Enfermedad Pulmonar Obstructiva Crónica/diagnóstico por imagen , Enfisema Pulmonar/diagnóstico por imagen , Cese del Hábito de Fumar , Adulto , Anciano , Estudios de Casos y Controles , Estudios de Cohortes , Progresión de la Enfermedad , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfisema Pulmonar/etiología , Enfisema Pulmonar/metabolismo , Fumar/efectos adversos , Fumar/metabolismo , Fumar/patología , Factores de Tiempo , Tomografía Computarizada por Rayos X
20.
Int Immunopharmacol ; 11(8): 1038-44, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21382533

RESUMEN

The role of tumor necrosis factor-α (TNF-α) in contributing to allergen induced airway remodeling in asthma is unknown. In this study we have utilized a mouse model of chronic OVA allergen induced airway remodeling to determine whether TNF p55/p75 receptor deficient mice (abbreviated TNF-R KO) had reduced levels of airway remodeling. Chronic OVA challenged WT mice had significantly increased levels of lung eosinophilic inflammation as well as features of airway remodeling including increased peribronchial fibrosis, thickness of the peribronchial smooth muscle layer, mucus expression, and deposition of extracellular matrix proteins. In contrast, TNF-R KO mice had significantly reduced levels of major basic protein positive peribronchial eosinophils and significantly reduced peribronchial fibrosis assessed by quantitating the area of peribronchial trichrome staining and total lung collagen. In addition, TNF-R KO mice had significantly reduced thickness of the peribronchial smooth muscle layer, area of peribronchial α-smooth muscle actin immunostaining, and levels of the extracellular matrix protein fibronectin. There was a non-significant trend for reduced mucus expression in TNF-R KO mice. Levels of peribronchial cells immunostaining positive for TGF-ß1 were significantly reduced in TNF-R KO mice suggesting that reduced levels of TGF-ß1 expression in TNF-R KO mice may contribute to reduced airway remodeling. Overall, this study suggests an important role for TNF-α in contributing to many features of allergen induced airway remodeling including changes in levels of peribronchial smooth muscle, subepithelial fibrosis, and deposition of extracellular matrix.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Alérgenos/farmacología , Ovalbúmina/farmacología , Eosinofilia Pulmonar/metabolismo , Eosinofilia Pulmonar/patología , Receptores Tipo II del Factor de Necrosis Tumoral/deficiencia , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Receptores Señuelo del Factor de Necrosis Tumoral/deficiencia , Animales , Asma/metabolismo , Asma/patología , Bronquios/metabolismo , Bronquios/patología , Colágeno/metabolismo , Eosinófilos/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Inmunoglobulina E/sangre , Interleucina-5/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Moco/metabolismo , Músculo Liso/efectos de los fármacos , Músculo Liso/inmunología , Músculo Liso/metabolismo , Músculo Liso/patología , Eosinofilia Pulmonar/inducido químicamente , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/metabolismo , Receptores Señuelo del Factor de Necrosis Tumoral/inmunología , Receptores Señuelo del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA