Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 713
Filtrar
1.
Respir Res ; 25(1): 227, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38812021

RESUMEN

BACKGROUND: Steroid insensitivity in Chronic Obstructive Pulmonary Disease (COPD) presents a problem for controlling the chronic inflammation of the airways. The glucocorticoid receptor (GR) mediates the intracellular signaling of inhaled corticosteroids (ICS) by interacting with transcription factors and histone deacetylases (HDACs). The aim of this study was to assess if COPD patients' response to ICS in vivo, may be associated with the expression of GR, the complex of GR with transcription factors, and the expression of various HDACs in vitro. METHODS: Primary airway smooth muscle cells (ASMC) were established from endobronchial biopsies obtained from patients with asthma (n = 10), patients with COPD (n = 10) and subjects that underwent diagnostic bronchoscopy without pathological findings and served as controls (n = 6). ASMC were also established from 18 COPD patients, 10 responders and 8 non-responders to ICS, who participated in the HISTORIC study, an investigator-initiated and driven clinical trial that proved the hypothesis that COPD patients with high ASMC in their endobronchial biopsies respond better to ICS than patients with low ASMC. Expression of GR and its isoforms GRα and GRß and HDACs was investigated in primary ASMC in the absence or in the presence of dexamethasone (10- 8M) by western blotting. The complex formation of GR with transcription factors was assessed by co-immunoprecipitation. RESULTS: Expression of GR and its isoform GRα but not GRß was significantly reduced in ASMC from COPD patients as compared to controls. There were no significant differences in the expression of GR, GRα and GRß between responders and non-responders to ICS. However, treatment with dexamethasone upregulated the expression of total GR (p = 0.004) and GRα (p = 0.005) after 30 min in responders but not in non-responders. Τhe formation of the complex GR-c-Jun was increased 60 min after treatment with dexamethasone only in responders who exhibited significantly lower expression of HDAC3 (p = 0.005) and HDAC5 (p < 0.0001) as compared to non-responders. CONCLUSIONS: These data suggest that ASMC from COPD patients who do not respond to treatment with ICS, are characterized by reduced GR-c-Jun complex formation and increased expression of HDAC3 and HDAC5. TRIAL REGISTRATION: ISRCTN11017699 (Registration date: 15/11/2016).


Asunto(s)
Histona Desacetilasas , Miocitos del Músculo Liso , Enfermedad Pulmonar Obstructiva Crónica , Receptores de Glucocorticoides , Humanos , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/patología , Receptores de Glucocorticoides/metabolismo , Receptores de Glucocorticoides/biosíntesis , Histona Desacetilasas/metabolismo , Histona Desacetilasas/biosíntesis , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Masculino , Persona de Mediana Edad , Femenino , Anciano , Células Cultivadas , Corticoesteroides/uso terapéutico , Glucocorticoides/farmacología , Dexametasona/farmacología , Resultado del Tratamiento , Administración por Inhalación , Bronquios/efectos de los fármacos , Bronquios/metabolismo , Bronquios/patología , Bronquios/enzimología
2.
Ecotoxicol Environ Saf ; 236: 113486, 2022 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-35397445

RESUMEN

Methanethiol is a widely existing malodorous pollutant with health effects on the human population. However, the cytotoxicity mechanism of methanethiol in vitro and its metabolic transformation (bioactivation or detoxification) have not been fully elucidated. Herein, the metabolites of methanethiol during cell culture and the cytotoxicity of methanethiol in human bronchial epithelial (16HBE) cells were investigated. Results indicate that methanethiol (10-50 µM) was partially converted into dimethyl sulfide, mainly catalyzed by thiol S-methyltransferase in the 16HBE cells, and then it induced potent cytotoxicity and cell membrane permeability. Moreover, methanethiol induced intracellular reactive oxygen species (ROS) up to 50 µM and further activated the tumor necrosis factor (TNF) signaling pathway, which eventually led to the decline in the mitochondrial membrane potential (MMP) and cell necrosis. However, all these effects were significantly alleviated with gene silencing of the methyltransferase-like protein 7B (METTL7B). These results indicate that methanethiol may induce cell necrosis in human respiratory tract cells mainly mediated by S-methyltransferase with interfering TNF and ROS induction. Non-target metabolomics results suggest that methanethiol potently affects expression of endogenous small molecule metabolites in 16HBE cells. To some extent, this work shows the possible conversion path and potential injury mechanism of human respiratory tract cells exposed to methanethiol.


Asunto(s)
Bronquios , Proteínas Portadoras , Metiltransferasas , Compuestos de Sulfhidrilo , Bronquios/efectos de los fármacos , Bronquios/enzimología , Bronquios/patología , Proteínas Portadoras/metabolismo , Activación Enzimática , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Células Epiteliales/patología , Humanos , Metiltransferasas/genética , Metiltransferasas/metabolismo , Necrosis , Especies Reactivas de Oxígeno/metabolismo , Compuestos de Sulfhidrilo/farmacología
3.
Int J Mol Sci ; 22(23)2021 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-34884593

RESUMEN

Subepithelial fibrosis is a component of the remodeling observed in the bronchial wall of patients diagnosed with asthma. In this process, human bronchial fibroblasts (HBFs) drive the fibroblast-to-myofibroblast transition (FMT) in response to transforming growth factor-ß1 (TGF-ß1), which activates the canonical Smad-dependent signaling. However, the pleiotropic properties of TGF-ß1 also promote the activation of non-canonical signaling pathways which can affect the FMT. In this study we investigated the effect of p38 mitogen-activated protein kinase (MAPK) inhibition by SB203580 on the FMT potential of HBFs derived from asthmatic patients using immunocytofluorescence, real-time PCR and Western blotting methods. Our results demonstrate for the first time the strong effect of p38 MAPK inhibition on the TGF-ß1-induced FMT potential throughout the strong attenuation of myofibroblast-related markers: α-smooth muscle actin (α-SMA), collagen I, fibronectin and connexin 43 in HBFs. We suggest the pleiotropic mechanism of SB203580 on FMT impairment in HBF populations by the diminishing of TGF-ß/Smad signaling activation and disturbances in the actin cytoskeleton architecture along with the maturation of focal adhesion sites. These observations justify future research on the role of p38 kinase in FMT efficiency and bronchial wall remodeling in asthma.


Asunto(s)
Asma/tratamiento farmacológico , Bronquios/efectos de los fármacos , Diferenciación Celular , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de los fármacos , Imidazoles/farmacología , Piridinas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Adulto , Asma/enzimología , Asma/patología , Bronquios/enzimología , Células Cultivadas , Femenino , Fibroblastos/enzimología , Humanos , Masculino , Persona de Mediana Edad , Transducción de Señal
4.
Immunol Lett ; 237: 33-41, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34228987

RESUMEN

OBJECTIVE: In this study, we focused on the interaction between SARS-CoV-2 and host Type I Interferon (IFN) response, so as to identify whether IFN effects could be influenced by the products of SARS-CoV-2. METHODS: All the structural and non-structural proteins of SARS-CoV-2 were transfected and overexpressed in the bronchial epithelial cell line BEAS-2B respectively, and typical antiviral IFN-stimulated gene (ISG) ISG15 expression was detected by qRT-PCR. RNA-seq based transcriptome analysis was performed between control and Spike (S) protein-overexpressed BEAS-2B cells. The expression of ACE2 and IFN effector JAK-STAT signaling activation were detected in control and S protein-overexpressed BEAS-2B cells by qRT-PCR or/and Western blot respectively. The interaction between S protein with STAT1 and STAT2, and the association between JAK1 with downstream STAT1 and STAT2 were measured in BEAS-2B cells by co-immunoprecipitation (co-IP). RESULTS: S protein could activate IFN effects and downstream ISGs expression. By transcriptome analysis, overexpression of S protein induced a set of genes expression, including series of ISGs and the SARS-CoV-2 receptor ACE2. Mechanistically, S protein enhanced the association between the upstream JAK1 and downstream STAT1 and STAT2, so as to promote STAT1 and STAT2 phosphorylation and ACE2 expression. CONCLUSION: SARS-CoV-2 S protein enhances ACE2 expression via facilitating IFN effects, which may help its infection.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Bronquios/efectos de los fármacos , COVID-19/virología , Células Epiteliales/efectos de los fármacos , Interferón alfa-2/farmacología , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Bronquios/enzimología , Bronquios/virología , COVID-19/enzimología , Citocinas/genética , Citocinas/metabolismo , Células Epiteliales/enzimología , Células Epiteliales/virología , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Janus Quinasa 1/metabolismo , Fosforilación , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Transducción de Señal , Glicoproteína de la Espiga del Coronavirus/genética , Ubiquitinas/genética , Ubiquitinas/metabolismo , Regulación hacia Arriba
5.
J Pathol ; 253(1): 17-30, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32991738

RESUMEN

Angiotensin-converting enzyme 2 (ACE2) has been identified as the functional receptor of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and a target for disease prevention. However, the relationship between ACE2 expression and its clinical implications in SARS-CoV-2 pathogenesis remains unknown. Here, we explored the location and expression of ACE2, and its correlation with gender, age, and cigarette smoke (CS), in a CS-exposed mouse model and 224 non-malignant lung tissues (125 non-smokers, 81 current smokers, and 18 ex-smokers) by immunohistochemistry. Moreover, the correlations of ACE2 with CS-induced oxidative stress-related markers, hypoxia-inducible factor-1α (HIF-1α), inducible nitric oxide synthase (iNOS), and 4-hydroxynonenal (4-HNE) were investigated. Chromatin immunoprecipitation and luciferase reporter assays identified the cause of ACE2 overexpression in human primary lung epithelial cells. We demonstrated that ACE2 was predominantly overexpressed on the apical surface of bronchial epithelium, while reduced in alveolar epithelium, owing to the dramatically decreased abundance of alveolar type II pneumocytes in CS-exposed mouse lungs. Consistent with this, ACE2 was primarily significantly overexpressed in human bronchial and alveolar epithelial cells in smokers regardless of age or gender. Decreased ACE2 expression was observed in bronchial epithelial cells from ex-smokers compared with current smokers, especially in those who had ceased smoking for more than 10 years. Moreover, ACE2 expression was positively correlated with the levels of HIF-1α, iNOS, and 4-HNE in both mouse and human bronchioles. The results were further validated using a publicly available dataset from The Cancer Genome Atlas (TCGA) and our previous integrated data from Affymetrix U133 Plus 2.0 microarray (AE-meta). Finally, our results showed that HIF-1α transcriptionally upregulates ACE2 expression. Our results indicate that smoking-induced ACE2 overexpression in the apical surface of bronchial epithelial cells provides a route by which SARS-CoV-2 enters host cells, which supports clinical relevance in attenuating the potential transmission risk of COVID-19 in smoking populations by smoking cessation. © 2020 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Células Epiteliales Alveolares/enzimología , Enzima Convertidora de Angiotensina 2/metabolismo , Bronquios/enzimología , COVID-19/virología , Células Epiteliales/virología , Fumar/efectos adversos , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Células Epiteliales Alveolares/virología , Animales , Niño , Preescolar , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Femenino , Humanos , Lactante , Pulmón/metabolismo , Pulmón/virología , Persona de Mediana Edad , SARS-CoV-2 , Adulto Joven
6.
Artículo en Inglés | MEDLINE | ID: mdl-32673988

RESUMEN

Pulmonary hypertension (PH) is a progressive and life-threating lung disorder characterized by elevated pulmonary artery pressure and vascular remodeling. PH is classified into five groups, and one of the most common and lethal forms, PH Group-III is defined as PH due to lung diseases and/or hypoxia. Due to the lack of studies in this group, PH-specific drug therapies including prostacyclin (PGI2) analogues have not been approved or recommended for use in these patients. PGI2 is synthesized by the PGI2 synthase (PGIS) enzyme, and its production is determined by measuring its stable metabolite, 6-keto-PGF1α. An impaired PGI2 pathway has been observed in PH animal models and in PH Group-I patients; however, there are contradictory results. The aim of this study is to determine whether PH Group-III is associated with altered expression of PGIS and production of PGI2 in humans. To explore this hypothesis, we measured PGIS expression (by western blot) and PGI2 production (by ELISA) in a large variety of preparations from the pulmonary circulation including human pulmonary artery, pulmonary vein, distal lung tissue, pulmonary artery smooth muscle cells (hPASMC), and bronchi in PH Group-III (n = 35) and control patients (n = 32). Our results showed decreased PGIS expression and/or 6-keto-PGF1α levels in human pulmonary artery, hPASMC, and distal lung tissue derived from PH Group-III patients. Moreover, the production of 6-keto-PGF1α from hPASMC positively correlated with PGIS expression and was inversely correlated with mean pulmonary artery pressure. On the other hand, PH Group-III pulmonary veins and bronchi did not show altered PGI2 production compared to controls. The deficit in PGIS expression and/or PGI2 production observed in pulmonary artery and distal lung tissue in PH Group-III patients may have important implications in the pathogenesis and treatment of PH Group-III.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Epoprostenol/metabolismo , Hipertensión Pulmonar/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Arteria Pulmonar/metabolismo , Bronquios/enzimología , Bronquios/metabolismo , Hipoxia de la Célula/fisiología , Células Cultivadas , Dinoprost/metabolismo , Regulación hacia Abajo , Femenino , Humanos , Hipertensión Pulmonar/enzimología , Hipertensión Pulmonar/fisiopatología , Pulmón/enzimología , Pulmón/metabolismo , Masculino , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/metabolismo , Arteria Pulmonar/enzimología , Venas Pulmonares/enzimología , Venas Pulmonares/metabolismo
7.
Respir Res ; 21(1): 188, 2020 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-32677970

RESUMEN

BACKGROUND: A disintegrin and metalloproteinase domain-15 (ADAM15) is expressed by activated leukocytes, and fibroblasts in vitro. Whether ADAM15 expression is increased in the lungs of COPD patients is not known. METHODS: ADAM15 gene expression and/or protein levels were measured in whole lung and bronchoalveolar lavage (BAL) macrophage samples obtained from COPD patients, smokers, and non-smokers. Soluble ADAM15 protein levels were measured in BAL fluid (BALF) and plasma samples from COPD patients and controls. Cells expressing ADAM15 in the lungs were identified using immunostaining. Staining for ADAM15 in different cells in the lungs was related to forced expiratory volume in 1 s (FEV1), ratio of FEV1 to forced vital capacity (FEV1/FVC), and pack-years of smoking history. RESULTS: ADAM15 gene expression and/or protein levels were increased in alveolar macrophages and whole lung samples from COPD patients versus smokers and non-smokers. Soluble ADAM15 protein levels were similar in BALF and plasma samples from COPD patients and controls. ADAM15 immunostaining was increased in macrophages, CD8+ T cells, epithelial cells, and airway α-smooth muscle (α-SMA)-positive cells in the lungs of COPD patients. ADAM15 immunostaining in macrophages, CD8+ T cells and bronchial (but not alveolar) epithelial cells was related inversely to FEV1 and FEV1/FVC, but not to pack-years of smoking history. ADAM15 staining levels in airway α-SMA-positive cells was directly related to FEV1/FVC. Over-expressing ADAM15 in THP-1 cells reduced their release of matrix metalloproteinases and CCL2. CONCLUSIONS: These results link increased ADAM15 expression especially in lung leukocytes and bronchial epithelial cells to the pathogenesis of COPD.


Asunto(s)
Proteínas ADAM/metabolismo , Bronquios/enzimología , Linfocitos T CD8-positivos/enzimología , Células Epiteliales/enzimología , Macrófagos Alveolares/enzimología , Proteínas de la Membrana/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/enzimología , Adulto , Anciano , Anciano de 80 o más Años , Beijing , Biomarcadores/metabolismo , Boston , Bronquios/fisiopatología , Estudios de Casos y Controles , Inglaterra , Femenino , Volumen Espiratorio Forzado , Humanos , Masculino , Persona de Mediana Edad , No Fumadores , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Fumadores , Células THP-1 , Regulación hacia Arriba , Capacidad Vital , Adulto Joven
8.
Glycoconj J ; 37(5): 623-633, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32666337

RESUMEN

Human primary bronchial epithelial cells differentiated in vitro represent a valuable tool to study lung diseases such as cystic fibrosis (CF), an inherited disorder caused by mutations in the gene coding for the Cystic Fibrosis Transmembrane Conductance Regulator. In CF, sphingolipids, a ubiquitous class of bioactive lipids mainly associated with the outer layer of the plasma membrane, seem to play a crucial role in the establishment of the severe lung complications. Nevertheless, no information on the involvement of sphingolipids and their metabolism in the differentiation of primary bronchial epithelial cells are available so far. Here we show that ceramide and globotriaosylceramide increased during cell differentiation, whereas glucosylceramide and gangliosides content decreased. In addition, we found that apical plasma membrane of differentiated bronchial cells is characterized by a higher content of sphingolipids in comparison to the other cell membranes and that activity of sphingolipids catabolic enzymes associated with this membrane results altered with respect to the total cell activities. In particular, the apical membrane of CF cells was characterized by high levels of ceramide and glucosylceramide, known to have proinflammatory activity. On this basis, our data further support the role of sphingolipids in the onset of CF lung pathology.


Asunto(s)
Diferenciación Celular/genética , Fibrosis Quística/genética , Hidrolasas/genética , Esfingolípidos/genética , Bronquios/enzimología , Membrana Celular/enzimología , Membrana Celular/genética , Ceramidas/genética , Fibrosis Quística/enzimología , Fibrosis Quística/metabolismo , Fibrosis Quística/patología , Glucosilceramidas/genética , Humanos , Hidrolasas/química , Cultivo Primario de Células , Esfingolípidos/metabolismo
9.
Expert Rev Respir Med ; 14(5): 501-510, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32098546

RESUMEN

Introduction: Mitogen-activated protein kinases (MAPKs) are a large family of evolutionary conserved intracellular enzymes that play a pivotal role in signaling pathways mediating the biologic actions of a wide array of extracellular stimuli.Areas covered: MAPKs are implicated in most pathogenic events involved in asthma, including both inflammatory and structural changes occurring in the airways. Indeed, MAPKs are located at the level of crucial convergence points within the signal transduction networks activated by many cytokines, chemokines, growth factors, and other inducers of bronchial inflammation and remodeling such as immunoglobulin E (IgE) and oxidative stress.Expert opinion: Therefore, given the growing importance of MAPKs in asthma pathobiology, these signaling enzymes are emerging as key intracellular pathways whose upstream activation can be inhibited by biological drugs such as anti-cytokines and anti-IgE.


Asunto(s)
Asma/enzimología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Asma/inmunología , Asma/metabolismo , Bronquios/enzimología , Bronquios/metabolismo , Citocinas/inmunología , Humanos , Estrés Oxidativo , Transducción de Señal
10.
BMC Pulm Med ; 19(1): 218, 2019 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-31747880

RESUMEN

BACKGROUND: The dysfunction of airway epithelial barrier is closely related to the pathogenesis of asthma. Secreted Hsp90α participates in inflammation and Hsp90 inhibitor protects endothelial dysfunction. In the current study, we aimed to explore the role of secreted Hsp90α in asthmatic airway epithelial barrier function. METHODS: Male BALB/c mice were sensitized and challenged with HDM to generate asthma model. The 16HBE and Hsp90α-knockdown cells were cultured and treated according to the experiment requirements. Transepithelial Electric Resistance (TEER) and permeability of epithelial layer in vitro, distribution and expression of junction proteins both in vivo and in vitro were used to evaluate the epithelial barrier function. Western Blot was used to evaluate the expression of junction proteins and phosphorylated AKT in cells and lung tissues while ELISA were used to evaluate the Hsp90α expression and cytokines release in the lung homogenate. RESULTS: HDM resulted in a dysfunction of airway epithelial barrier both in vivo and in vitro, paralleled with the increased expression and release of Hsp90α. All of which were rescued in Hsp90α-knockdown cells or co-administration of 1G6-D7. Furthermore, either 1G6-D7 or PI3K inhibitor LY294002 suppressed the significant phosphorylation of AKT, which caused by secreted and recombinant Hsp90α, resulting in the restoration of epithelial barrier function. CONCLUSIONS: Secreted Hsp90α medicates HDM-induced asthmatic airway epithelial barrier dysfunction via PI3K/AKT pathway, indicating that anti-secreted Hsp90α therapy might be a potential treatment to asthma in future.


Asunto(s)
Asma/fisiopatología , Bronquios/efectos de los fármacos , Cromonas/farmacología , Células Epiteliales/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/metabolismo , Morfolinas/farmacología , Animales , Asma/tratamiento farmacológico , Bronquios/enzimología , Bronquios/inmunología , Cadherinas/metabolismo , Línea Celular , Citocinas/metabolismo , Impedancia Eléctrica , Células Epiteliales/enzimología , Células Epiteliales/inmunología , Técnicas de Silenciamiento del Gen , Proteínas HSP90 de Choque Térmico/genética , Humanos , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Fosforilación , Pyroglyphidae/inmunología , Mucosa Respiratoria/metabolismo
11.
Redox Biol ; 26: 101256, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31229842

RESUMEN

Environmental proteases have been widely associated to the pathogenesis of allergic disorders. Der p 1, a cysteine-protease from house dust mite (HDM) Dermatophagoides pteronyssinus, constitutes one of the most clinically relevant indoor aeroallergens worldwide. Der p 1 protease activity depends on the redox status of its catalytic cysteine residue, which has to be in the reduced state to be active. So far, it is unknown whether Der p 1-protease activity could be regulated by host redox microenvironment once it reaches the lung epithelial lining fluid in addition to endogenous mite components. In this sense, Glutathione-S-transferase pi (GSTpi), an enzyme traditionally linked to phase II detoxification, is highly expressed in human lung epithelial cells, which represent the first line of defence against aeroallergens. Moreover, GSTpi is a generalist catalyst of protein S-glutathionylation reactions, and some polymorphic variants of this enzyme has been associated to the development of allergic asthma. Here, we showed that human GSTpi increased the cysteine-protease activity of Der p 1, while GSTmu (the isoenzyme produced by the mite) did not alter it. GSTpi induces the reduction of Cys residues in Der p 1, probably by rearranging its disulphide bridges. Furthermore, GSTpi was detected in the apical medium collected from human bronchial epithelial cell cultures, and more interesting, it increased cysteine-protease activity of Der p 1. Our findings support the role of human GSTpi from airways in modulating of Der p 1 cysteine-protease activity, which may have important clinical implications for immune response to this aeroallergen in genetically susceptible individuals.


Asunto(s)
Antígenos Dermatofagoides/metabolismo , Proteínas de Artrópodos/metabolismo , Cisteína Endopeptidasas/metabolismo , Cisteína/metabolismo , Dermatophagoides pteronyssinus/química , Células Epiteliales/enzimología , Gutatión-S-Transferasa pi/metabolismo , Animales , Antígenos Dermatofagoides/inmunología , Proteínas de Artrópodos/inmunología , Bronquios/citología , Bronquios/enzimología , Bronquios/inmunología , Línea Celular , Cisteína/inmunología , Cisteína Endopeptidasas/inmunología , Dermatophagoides pteronyssinus/enzimología , Dermatophagoides pteronyssinus/inmunología , Células Epiteliales/citología , Células Epiteliales/inmunología , Gutatión-S-Transferasa pi/inmunología , Humanos , Isoenzimas/inmunología , Isoenzimas/metabolismo , Cinética , Oxidación-Reducción , Proteolisis , Especificidad de la Especie
12.
Food Chem Toxicol ; 127: 89-100, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30849403

RESUMEN

The aim of this study was to evaluate cytotoxicity (WST-1 assay), LDH release (LDH assay) and genotoxicity (Comet assay) of three engineered TiO2-NPs with different shapes (bipyramids, rods, platelets) in comparison with two commercial TiO2-NPs (P25, food grade). After NPs characterization (SEM/T-SEM and DLS), biological effects of NPs were assessed on BEAS-2B cells in presence/absence of light. The cellular uptake of NPs was analyzed using Raman spectroscopy. The cytotoxic effects were mostly slight. After light exposure, the largest cytotoxicity (WST-1 assay) was observed for rods; P25, bipyramids and platelets showed a similar effect; no effect was induced by food grade. No LDH release was detected, confirming the low effect on plasma membrane. Food grade and platelets induced direct genotoxicity while P25, food grade and platelets caused oxidative DNA damage. No genotoxic or oxidative damage was induced by bipyramids and rods. Biological effects were overall lower in darkness than after light exposure. Considering that only food grade, P25 and platelets (more agglomerated) were internalized by cells, the uptake resulted correlated with genotoxicity. In conclusion, cytotoxicity of NPs was low and affected by shape and light exposure, while genotoxicity was influenced by cellular-uptake and aggregation tendency.


Asunto(s)
Bronquios/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayo Cometa , Células Epiteliales/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Titanio/toxicidad , Bronquios/citología , Bronquios/enzimología , Línea Celular , Daño del ADN , Células Epiteliales/enzimología , Humanos , L-Lactato Deshidrogenasa/metabolismo , Microscopía Electrónica de Transmisión de Rastreo , Estrés Oxidativo/efectos de los fármacos , Tamaño de la Partícula , Espectrometría Raman/métodos
13.
Toxicol Sci ; 169(1): 224-234, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30715550

RESUMEN

Chronic exposure of human bronchial epithelial BEAS-2B cells to hexavalent chromium [Cr(VI)] causes malignant cell transformation. Sirtuin-3 (SIRT3) regulates mitochondrial adaptive response to stress, such as metabolic reprogramming and antioxidant defense mechanisms. In Cr(VI)-transformed cells, SIRT3 was upregulated and mitochondrial adenosine triphosphate (ATP) production and proton leak were reduced. Knockdown of SIRT3 by its shRNA further decreased mitochondrial ATP production, proton leak, mitochondrial mass, and mitochondrial membrane potential, indicating that SIRT3 positively regulates mitochondrial oxidative phosphorylation and maintenance of mitochondrial integrity. Mitophagy is critical to maintain proper cellular functions. In Cr(VI)-transformed cells expressions of Pink 1 and Parkin, two mitophagy proteins, were elevated, and mitophagy remained similar as that in passage-matched normal BEAS-2B cells, indicating that in -Cr(VI)-transformed cells mitophagy is suppressed. Knockdown of SIRT3 induced mitophagy, suggesting that SIRT3 plays an important role in mitophagy suppression of Cr(VI)-transformed cells. In Cr(VI)-transformed cells, nuclear factor (erythroid-derived 2)-like 2 (Nrf2) was constitutively activated, and protein levels of p62 and p-p62Ser349 were elevated. Knockdown of SIRT3 or treatment with carbonyl cyanide m-chlorophenyl hydrazone (CCCP) decreased the binding of p-p62Ser349 to Keap1, resulting in increased binding of Keap1 to Nrf2 and consequently reduced Nrf2 activation. The results from CHIP assay showed that in Cr(VI)-transformed cells binding of Nrf2 to antioxidant response element (ARE) of SIRT3 gene promoter was dramatically increased. Knockdown of SIRT3 suppressed cell proliferation and tumorigenesis of Cr(VI)-transformed cells. Overexpression of SIRT3 in normal BEAS-2B cells exhibited mitophagy suppression phenotype and increased cell proliferation and tumorigenesis. The present study demonstrated that upregulation of SIRT3 causes mitophagy suppression and plays an important role in cell survival and tumorigenesis of Cr(VI)-transformed cells.


Asunto(s)
Bronquios/efectos de los fármacos , Transformación Celular Neoplásica/inducido químicamente , Cromatos/toxicidad , Cromo/toxicidad , Células Epiteliales/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Sirtuina 3/metabolismo , Bronquios/enzimología , Bronquios/patología , Línea Celular Transformada , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Metabolismo Energético/efectos de los fármacos , Activación Enzimática , Células Epiteliales/enzimología , Células Epiteliales/patología , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Mitocondrias/enzimología , Mitocondrias/genética , Mitocondrias/patología , Mitofagia/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Proteína Sequestosoma-1/metabolismo , Transducción de Señal , Sirtuina 3/genética
14.
Arch Biochem Biophys ; 664: 167-173, 2019 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-30677406

RESUMEN

Human airway trypsin-like protease (HAT) localizes at human bronchial epithelial cells (HBECs). HAT enhanced release of interleukin-8 (IL-8) from HBECs at 10-100 mU/mL and the enhanced release was almost completely abolished by 50 µM leupeptin, a serine protease inhibitor. Previous reports suggested that HAT displays its physiological functions via protease-activated receptor 2 (PAR2). In the present study, we examined the mechanism whereby HAT upregulates IL-8 synthesis in HBECs with a focus on PAR2. Northern blot analysis revealed that HAT enhanced IL-8 mRNA expression at concentrations of 10-100 mU/mL. PAR2 activating peptide (PAR2 AP) also enhanced IL-8 release and IL-8 mRNA expression in HBECs at 50-1,000 µM at similar levels as HAT. Knockdown of PAR2 mRNA by siRNA methods showed that PAR2 mRNA expression was significantly depressed in primary HBECs, and both HAT- and PAR2 AP-induced IL-8 mRNA elevation was significantly depressed in PAR2 siRNA-transfected HBECs. Additionally, HAT cleaved the PAR2 activating site (R36-S37 bond) of synthetic PAR2 N-terminal peptide. These results indicate that HAT stimulates IL-8 synthesis in airway epithelial cells via PAR2 and could help to amplify inflammation in chronic respiratory tract disease.


Asunto(s)
Bronquios/enzimología , Interleucina-8/biosíntesis , Receptor PAR-2/metabolismo , Serina Endopeptidasas/metabolismo , Secuencia de Aminoácidos , Bronquios/citología , Células Cultivadas , Células Epiteliales/metabolismo , Humanos , Interleucina-8/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética
15.
Biosci Rep ; 39(1)2019 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-30573530

RESUMEN

Mycoplasma pneumoniae is one of the most common pathogenic causes of community-acquired pneumonia. Hydrogen sulfide, alanine, and pyruvate producing enzyme (HapE) is a recently discovered M. pneumoniae virulence factor that can produce H2S to promote erythrocyte lysis. However, other cytotoxic effects of HapE have not been explored. The present study examined the effects of this enzyme on normal human bronchial epithelial (NHBE) cells, in an attempt to identify additional mechanisms of M. pneumoniae pathogenesis. Recombinant HapE was purified for use in downstream assays. MTT and colony formation assays were conducted to determine the effects of HapE on cell viability and growth, while flow cytometry was used to examine changes in cell proliferation and cell cycle function. ELISA was performed to examine changes in the cytokine profile of HapE-treated cells. HapE treatment arrested NHBE cells in S phase and inhibited cell proliferation in a concentration-dependent manner. The anti-inflammatory factors interleukin (IL)-4 and IL-6 were significantly enhanced following HapE treatment. Increased secretion of pro-inflammatory factors was not observed. The effects of HapE on the respiratory epithelium may have an impact on the efficiency of host immune surveillance and pathogen elimination, and contribute to the pathogenesis of M. pneumoniae.


Asunto(s)
Proteínas Bacterianas/genética , Bronquios/microbiología , Mycoplasma pneumoniae/enzimología , Neumonía por Mycoplasma/enzimología , Factores de Virulencia/genética , Proteínas Bacterianas/farmacología , Bronquios/enzimología , Proliferación Celular/genética , Citocinas/genética , Citocinas/farmacología , Células Epiteliales/enzimología , Células Epiteliales/microbiología , Humanos , Mycoplasma pneumoniae/genética , Mycoplasma pneumoniae/patogenicidad , Neumonía por Mycoplasma/genética , Neumonía por Mycoplasma/microbiología , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Factores de Virulencia/farmacología
16.
J Cell Physiol ; 233(12): 9763-9776, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30078190

RESUMEN

Primary porcine bronchial epithelial cells (PBECs) are an ideal model to study the molecular and pathogenic mechanisms of various porcine respiratory pathogens. However, the short lifespan of primary PBECs greatly limit their application. Here, we isolated and cultured primary PBECs and established immortalized PBECs by transfecting primary PBECs with the pEGFP-hTERT recombinant plasmid containing human telomerase reverse transcriptase (hTERT). Immortalized PBECs (hTERT-PBECs) retained the morphological and functional features of primary PBECs as indicated by cytokeratin 18 expression, telomerase activity assay, proliferation assays, karyotype analysis, and quantitative reverse-transcriptase polymerase chain reaction. Compared to primary PBECs, hTERT-PBECs had higher telomerase activity, extended replicative lifespan, and displayed enhanced proliferative activity. Moreover, this cell line is not transformed in vitro and does not exhibit a malignant phenotype in vivo, suggesting that it can be safely used in further studies. Besides, hTERT-PBECs were susceptible to swine influenza virus of H3N2 subtype and porcine circovirus type 2. In conclusion, the immortalized hTERT-PBECs represent a valuable in vitro model, which can be widely used in the study of porcine respiratory pathogenic infections.


Asunto(s)
Bronquios/citología , Células Epiteliales/enzimología , Cultivo Primario de Células/métodos , Telomerasa/genética , Animales , Bronquios/enzimología , Proliferación Celular/genética , Circovirus/patogenicidad , Humanos , Cariotipo , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/virología , Porcinos , Telomerasa/biosíntesis
17.
Toxicol Lett ; 295: 270-276, 2018 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-29981922

RESUMEN

Poly (ADP-ribosylation) is a key post-translational modification (PTM), and poly (ADP-ribose) glycohydrolase (PARG) is the main enzyme that hydrolyzes poly (ADP-ribose) in eukaryotic organisms. Our previous findings suggested that knockdown of PARG attenuates benzo(a)pyrene (BaP) carcinogenesis. However, the mechanisms underlying PARG-mediated protective effects remain limited. In this study, the expression levels of histones were analyzed by Western blotting and immunofluorescence. Histone H2A levels were abnormally decreased by BaP-induced carcinogenesis, but were maintained by knockdown of PARG in the 16HBE human bronchial epithelial cell line. The interaction between poly (ADP-ribose) and H2A was confirmed by co-immunoprecipitation. PARG-related modifications in H2A were profiled by immune antibody enrichment coupled with mass spectrometry. H2AK5ac, H2AK9ac, H2AK13ac, H2A.ZK4K7K11ac, and H2AK9me were expressed in BaP-transformed 16HBE (BTC-16HBE) cells, but were not detectable in normal 16HBE or BaP-transformed 16HBE cells with knockdown of PARG (BTC-shPARG). Further verification by Western blotting indicated that H2AK9me was elevated in BTC-16HBE cells but decreased in BTC-shPARG cells. These findings suggest that knockdown of PARG protects against BaP-induced carcinogenesis in 16HBE cells by downregulating H2AK9me. Our in vivo studies confirmed that PARG silencing decreased H2AK9me levels, thereby countering the carcinogenic teratogenic effects induced by BaP.


Asunto(s)
Benzo(a)pireno/toxicidad , Bronquios/efectos de los fármacos , Neoplasias de los Bronquios/prevención & control , Carcinógenos/toxicidad , Transformación Celular Neoplásica/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Glicósido Hidrolasas/metabolismo , Histonas/metabolismo , Interferencia de ARN , ADP-Ribosilación , Bronquios/enzimología , Bronquios/patología , Neoplasias de los Bronquios/inducido químicamente , Neoplasias de los Bronquios/enzimología , Neoplasias de los Bronquios/genética , Línea Celular , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Células Epiteliales/enzimología , Células Epiteliales/patología , Glicósido Hidrolasas/genética , Humanos
18.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L150-L156, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28982736

RESUMEN

The opportunistic pathogen Pseudomonas aeruginosa colonizes the lungs of susceptible individuals by deploying virulence factors targeting host defenses. The secreted factor Cif (cystic fibrosis transmembrane conductance regulator inhibitory factor) dysregulates the endocytic recycling of CFTR and thus reduces CFTR abundance in host epithelial membranes. We have postulated that the decrease in ion secretion mediated by Cif would slow mucociliary transport and decrease bacterial clearance from the lungs. To test this hypothesis, we explored the effects of Cif in cultured epithelia and in the lungs of mice. We developed a strategy to interpret the "hurricane-like" motions observed in reconstituted cultures and identified a Cif-mediated decrease in the velocity of mucus transport in vitro. Presence of Cif also increased the number of bacteria recovered at two time points in an acute mouse model of pneumonia caused by P. aeruginosa. Furthermore, recent work has demonstrated an inverse correlation between the airway concentrations of Cif and 15-epi-lipoxin A4, a proresolving lipid mediator important in host defense and the resolution of pathogen-initiated inflammation. Here, we observe elevated levels of 15-epi-lipoxin A4 in the lungs of mice infected with a strain of P. aeruginosa that expresses only an inactive form of cif compared with those mice infected with wild-type P. aeruginosa. Together these data support the inclusion of Cif on the list of virulence factors that assist P. aeruginosa in colonizing and damaging the airways of compromised patients. Furthermore, this study establishes techniques that enable our groups to explore the underlying mechanisms of Cif effects during respiratory infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bronquios/patología , Células Epiteliales/patología , Neumonía/etiología , Infecciones por Pseudomonas/complicaciones , Pseudomonas aeruginosa/patogenicidad , Factores de Virulencia/metabolismo , Animales , Transporte Biológico , Bronquios/enzimología , Bronquios/microbiología , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/enzimología , Células Epiteliales/microbiología , Humanos , Lipoxinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Depuración Mucociliar , Neumonía/metabolismo , Neumonía/patología , Infecciones por Pseudomonas/microbiología
19.
Am J Physiol Cell Physiol ; 314(3): C278-C288, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29141922

RESUMEN

receptor potential canonical (TRPC) channels are presently an emerging target for airway disorders. Recent evidence has indicated that TRPC6 as a member of the TRPC family plays an important role in airway inflammation, but its precise function in bronchial epithelial cells remains unclear. The aim of this study was to investigate the role of TRPC6 in Toll-like receptor 4 (TLR4)-mediated inflammation in human bronchial epithelial cells stimulated by endotoxin [lipopolysaccharide (LPS)]. Hyp9 is a simplified phloroglucinol derivative of hyperforin that highly selectively activates TRPC6 channels. The results show that the activation of TRPC6 by Hyp9 induced the production of interleukin (IL)-8 and IL-6. LPS was also able to induce the release of IL-8 and IL-6, which was significantly aggravated by Hyp9 and reduced by knockdown of TRPC6. Treatment with LPS not only chronically induced the expression of TRPC6 mRNA and protein in a TLR4-dependent manner but also acutely increased Ca2+ influx through TRPC6 channels. In addition, LPS-induced overexpression of TRPC6 and Ca2+ influx were associated with the phosphorylation of phosphatidylinositol 3-kinase (PI3K) and Akt. Importantly, TRPC6 was required for the activation of ERK1/2, p38, and NF-κB. In conclusion, these data reveal that LPS induced the overexpression of TRPC6 and TRPC6-dependent Ca2+ influx via the TLR4/PI3K/Akt pathway resulting in Ca2+ mobilization, which subsequently promoted the activation of ERK1/2, p38, and NF-κB and the inflammatory response in bronchial epithelial cells.


Asunto(s)
Bronquios/diagnóstico por imagen , Células Epiteliales/efectos de los fármacos , Inflamación/inducido químicamente , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Canal Catiónico TRPC6/agonistas , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Bronquios/enzimología , Señalización del Calcio/efectos de los fármacos , Línea Celular , Citocinas/metabolismo , Células Epiteliales/enzimología , Humanos , Inflamación/enzimología , Inflamación/genética , Mediadores de Inflamación/metabolismo , FN-kappa B/metabolismo , Floroglucinol/análogos & derivados , Floroglucinol/farmacología , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Canal Catiónico TRPC6/genética , Canal Catiónico TRPC6/metabolismo , Terpenos/farmacología , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/metabolismo
20.
Int J Chron Obstruct Pulmon Dis ; 12: 3341-3351, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29200841

RESUMEN

OBJECTIVE: To explore the effect of cigarette smoke (CS) on the development of squamous metaplasia in human airway epithelial cells and the role of MAPK- and FoxA2-signaling pathways in the process. MATERIALS AND METHODS: In an in vitro study, we treated the bronchial epithelial cell line BEAS2B with CS extract, followed by treatment with the ERK inhibitor U0126, the JNK inhibitor SP600125, or the p38 inhibitor SB203580. In vivo, we used a CS-induced rat model. After treatment with CS with or without MAPK inhibitors for 90 days, lung tissues were harvested. p-ERK, p-p38 and p-JNK protein levels in cells and lung tissue were measured using enzyme-linked immunosorbent assays, mRNA- and protein-expression profiles of FoxA2, E-cadherin, CD44, and ZO1 were measured using quantitative real-time polymerase chain reaction and Western blotting, respectively, and morphological changes in bronchial epithelial cells were observed using lung-tissue staining. RESULTS: In both the in vitro and in vivo studies, phosphorylation of the ERK1/2, JNK, and p38 proteins was significantly increased (P<0.05) and mRNA and protein expression of E-cadherin and FoxA2 significantly decreased (P<0.05) compared with the control group. ERK, JNK, and p38 inhibitors reversed the CS-extract-induced changes in E-cadherin, CD44, and ZO1 mRNA and protein expression (P<0.05), decreased p-ERK, p-p38, and p-JNK protein levels in cells and lung tissue, suppressed bronchial epithelial hyperplasia and local squamous metaplasia, and decreased FoxA2 expression. CONCLUSION: MAPK and FoxA2 mediate CS-induced squamous metaplasia. MAPK inhibitors upregulate FoxA2, resulting in a reduction in the degree of squamous metaplasia.


Asunto(s)
Bronquios/efectos de los fármacos , Fumar Cigarrillos/efectos adversos , Células Epiteliales/efectos de los fármacos , Factor Nuclear 3-beta del Hepatocito/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/etiología , Humo/efectos adversos , Animales , Bronquios/enzimología , Bronquios/patología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Fumar Cigarrillos/metabolismo , Fumar Cigarrillos/patología , Modelos Animales de Enfermedad , Células Epiteliales/enzimología , Células Epiteliales/patología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor Nuclear 3-beta del Hepatocito/genética , Humanos , Hiperplasia , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Metaplasia , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Enfermedad Pulmonar Obstructiva Crónica/enzimología , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfermedad Pulmonar Obstructiva Crónica/prevención & control , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA