Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
BMC Pulm Med ; 21(1): 98, 2021 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-33757467

RESUMEN

OBJECTIVE: Acute lung injury (ALI) is characterized by inflammation and oxidative stress. Nuclear factor-kappaB (NF-κB) mediates the expression of various inflammation-related genes, including the NADPH oxidase family. This study aimed to identify the potential regulatory role of NF-κB on NADPH oxidases in tumor necrosis factor-α (TNF-α)-induced oxidative stress in human alveolar epithelial cells. METHODS: A549 cells were treated with TNF-α for 24 h to establish ALI cell models. RT-PCR, western blot, assessment of oxidative stress, Alibaba 2.1 online analysis, electrophoretic mobility shift assays and luciferase reporter analysis were employed to identify the potential regulatory role of NF-κB on NADPH oxidases in TNF-α-induced oxidative stress in human alveolar epithelial cells. RESULTS: The expression of NF-κB/p65 was notably upregulated in TNF-α-stimulated A549 cells. NF-κB knockdown by siRNA significantly inhibited the TNF-α-induced oxidative stress. Moreover, NF-κB/p65 siRNA could inhibit the activation of NOX1, NOX2 and NOX4 mRNA and protein expression in TNF-α-stimulated A549 cells. The next study demonstrated that NF-κB activated the transcription of NOX1 by binding to the -261 to -252 bp (NOX1/κB2, TAAAAATCCC) region of NOX1 promoter in TNF-α-stimulated A549 cells. CONCLUSION: Our data demonstrated that NF-κB can aggravate TNF-α-induced ALI by regulating the oxidative stress response and the expression of NOX1, NOX2 and NOX4. Moreover, NF-κB could promote the NOX1 transcriptional activity via binding its promoter in TNF-α-stimulated A549 cells.


Asunto(s)
Lesión Pulmonar Aguda/enzimología , Células Epiteliales Alveolares/enzimología , NADPH Oxidasa 1/genética , NADPH Oxidasas/genética , FN-kappa B/metabolismo , Células A549 , Lesión Pulmonar Aguda/genética , Células Epiteliales Alveolares/efectos de los fármacos , Células Cultivadas , Regulación Enzimológica de la Expresión Génica , Humanos , Inflamación/enzimología , Inflamación/genética , Estrés Oxidativo/efectos de los fármacos , Transcripción Genética , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba
3.
Am J Respir Cell Mol Biol ; 62(6): 699-708, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32208980

RESUMEN

The mTOR pathway is one of the key signal cascades in the pathogenesis of idiopathic pulmonary fibrosis. Previous studies have mainly focused on this pathway in the fibroblasts and/or myofibroblasts, but not in the epithelial cells. In this study, we sought to investigate the role of the mTOR pathway in lung epithelial cells in lung fibrosis. Using Sftpc-mTORSL1+IT transgenic mice, in which active mTOR is conditionally expressed in lung epithelial cells, we assessed the effects of chronically activated mTOR in lung epithelial cells on lung phenotypes as well as bleomycin-induced lung fibrosis. Furthermore, we isolated alveolar epithelial cell type 2 from mice and performed RNA sequencing. Sftpc-mTORSL1+IT transgenic mice had no obvious abnormal findings, but, after bleomycin administration, showed more severe fibrotic changes and lower lung compliance than control mice. RNA sequencing revealed Angptl4 (angiopoietin-like protein 4) as a candidate downstream gene of the mTOR pathway. In vitro studies revealed that ANGPTL4, as well as mTOR, promoted tight junction vulnerability and epithelial-mesenchymal transition. mTOR activation in lung epithelial cells promoted lung fibrosis and the expression of ANGPTL4, a novel downstream target of the mTOR pathway, which could be related to the etiology of fibrosis.


Asunto(s)
Células Epiteliales Alveolares/enzimología , Transición Epitelial-Mesenquimal/fisiología , Fibrosis Pulmonar Idiopática/enzimología , Pulmón/enzimología , Serina-Treonina Quinasas TOR/fisiología , Células A549 , Células Epiteliales Alveolares/patología , Proteína 4 Similar a la Angiopoyetina/biosíntesis , Proteína 4 Similar a la Angiopoyetina/genética , Animales , Bleomicina/toxicidad , Caveolina 1/biosíntesis , Caveolina 1/genética , Activación Enzimática , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/patología , Pulmón/patología , Masculino , Ratones , Ratones Transgénicos , Fenotipo , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/genética , Proteína de la Zonula Occludens-1/biosíntesis , Proteína de la Zonula Occludens-1/genética
4.
Respir Res ; 21(1): 329, 2020 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-33308227

RESUMEN

AIMS: Acute lung injury (ALI) is a clinical syndrome with high morbidity and mortality, and severe pulmonary edema is one of the characteristics. Epithelial sodium channel (ENaC) located on the apical side of alveolar type 2 epithelial (AT2) cells is the primary rate limiting segment in alveolar fluid clearance. Many preclinical studies have revealed that mesenchymal stem cells (MSCs) based therapy has great therapeutic potential for ALI, while the role of ENaC in this process is rarely known. METHODS: We studied the effects of bone marrow-derived MSCs (BMSCs) on the protein/mRNA expression and activity of ENaC in primary mouse AT2 and human H441 cells by co-culture with them, respectively. Moreover, the changes of miRNA-130b in AT2 cells were detected by qRT-PCR, and we studied the involvement of phosphatase and tensin homolog deleted on chromosome ten (PTEN) and the downstream PI3K/AKT pathway in the miRNA-130b regulation of ENaC. RESULTS: Our results demonstrated that BMSCs could increase ENaC protein expression and function, as well as the expression level of miRNA-130b. The dual luciferase target gene assay verified that PTEN was one of the target genes of miR-130b, which showed adverse effects on the protein expression of α/γ-ENaC and PTEN in AT2 cells. Upregulating miR-130b and/or knocking down PTEN resulted in the increase of α/γ-ENaC protein level, and the protein expression of p-AKT/AKT was enhanced by miR-130b. Both α and γ-ENaC protein expressions were increased after AT2 cells were transfected with siPTEN, which could be reversed by the co-administration of PI3K/AKT inhibitor LY294002. CONCLUSION: In summary, miRNA-130b in BMSCs can enhance ENaC at least partially by targeting PTEN and activating PI3K/AKT pathway, which may provide a promising new direction for therapeutic strategy in ALI.


Asunto(s)
Células Epiteliales Alveolares/enzimología , Canales Epiteliales de Sodio/metabolismo , Neoplasias Pulmonares/enzimología , Células Madre Mesenquimatosas/metabolismo , MicroARNs/metabolismo , Fosfohidrolasa PTEN/metabolismo , Animales , Comunicación Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Canales Epiteliales de Sodio/genética , Neoplasias Pulmonares/genética , Masculino , Ratones Endogámicos C57BL , MicroARNs/genética , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo
5.
Cell Microbiol ; 21(10): e13085, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31290210

RESUMEN

Staphylococcus aureus is frequently isolated from patients with community-acquired pneumonia and acute respiratory distress syndrome (ARDS). ARDS is associated with staphylococcal phosphatidylinositol-specific phospholipase C (PI-PLC); however, the role of PI-PLC in the pathogenesis and progression of ARDS remains unknown. Here, we showed that recombinant staphylococcal PI-PLC possesses enzyme activity that causes shedding of glycosylphosphatidylinositol-anchored CD55 and CD59 from human umbilical vein endothelial cell surfaces and triggers cell lysis via complement activity. Intranasal infection with PI-PLC-positive S. aureus resulted in greater neutrophil infiltration and increased pulmonary oedema compared with a plc-isogenic mutant. Although indistinguishable proinflammatory genes were induced, the wild-type strain activated higher levels of C5a in lung tissue accompanied by elevated albumin instillation and increased lactate dehydrogenase release in bronchoalveolar lavage fluid compared with the plc- mutant. Following treatment with cobra venom factor to deplete complement, the wild-type strain with PI-PLC showed a reduced ability to trigger pulmonary permeability and tissue damage. PI-PLC-positive S. aureus induced the formation of membrane attack complex, mainly on type II pneumocytes, and reduced the level of CD55/CD59, indicating the importance of complement regulation in pulmonary injury. In conclusion, S. aureus PI-PLC sensitised tissue to complement activation leading to more severe tissue damage, increased pulmonary oedema, and ARDS progression.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas del Sistema Complemento/metabolismo , Fosfoinositido Fosfolipasa C/metabolismo , Edema Pulmonar/inmunología , Edema Pulmonar/microbiología , Síndrome de Dificultad Respiratoria/microbiología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/enzimología , Células Epiteliales Alveolares/enzimología , Células Epiteliales Alveolares/inmunología , Células Epiteliales Alveolares/microbiología , Animales , Proteínas Bacterianas/genética , Antígenos CD55/inmunología , Antígenos CD59/inmunología , Citocinas/metabolismo , Glicosilfosfatidilinositoles/inmunología , Glicosilfosfatidilinositoles/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos BALB C , Fosfoinositido Fosfolipasa C/genética , Edema Pulmonar/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/metabolismo , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo
6.
Int J Mol Sci ; 21(4)2020 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-32098115

RESUMEN

Alveolar edema, impaired alveolar fluid clearance, and elevated CO2 levels (hypercapnia) are hallmarks of the acute respiratory distress syndrome (ARDS). This study investigated how hypercapnia affects maturation of the Na,K-ATPase (NKA), a key membrane transporter, and a cell adhesion molecule involved in the resolution of alveolar edema in the endoplasmic reticulum (ER). Exposure of human alveolar epithelial cells to elevated CO2 concentrations caused a significant retention of NKA-ß in the ER and, thus, decreased levels of the transporter in the Golgi apparatus. These effects were associated with a marked reduction of the plasma membrane (PM) abundance of the NKA-α/ß complex as well as a decreased total and ouabain-sensitive ATPase activity. Furthermore, our study revealed that the ER-retained NKA-ß subunits were only partially assembled with NKA α-subunits, which suggests that hypercapnia modifies the ER folding environment. Moreover, we observed that elevated CO2 levels decreased intracellular ATP production and increased ER protein and, particularly, NKA-ß oxidation. Treatment with α-ketoglutaric acid (α-KG), which is a metabolite that has been shown to increase ATP levels and rescue mitochondrial function in hypercapnia-exposed cells, attenuated the deleterious effects of elevated CO2 concentrations and restored NKA PM abundance and function. Taken together, our findings provide new insights into the regulation of NKA in alveolar epithelial cells by elevated CO2 levels, which may lead to the development of new therapeutic approaches for patients with ARDS and hypercapnia.


Asunto(s)
Células Epiteliales Alveolares/enzimología , Dióxido de Carbono/metabolismo , Retículo Endoplásmico/enzimología , Hipercapnia/enzimología , Pliegue de Proteína , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Células A549 , Células Epiteliales Alveolares/patología , Animales , Retículo Endoplásmico/patología , Humanos , Hipercapnia/patología , Ratas
7.
J Stroke Cerebrovasc Dis ; 29(9): 104942, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32807413

RESUMEN

BACKGROUND AND OBJECTIVES: Studies implicate the lung in moderating systemic immune activation via effects on circulating leukocytes. In this study, we investigated whether targeted expression of the antioxidant extracellular superoxide dismutase (SOD3) within the lung would influence post-ischemic peripheral neutrophil activation and CNS reperfusion injury. METHODS: Adult, male mice expressing human SOD3 within type II pneumocytes were subjected to 15 min of transient global cerebral ischemia. Three days post-reperfusion, lung and brain tissue was collected and analyzed by immunohistochemistry for inflammation and injury markers. In vitro motility and neurotoxicity assays were conducted to ascertain the direct effects of hSOD3 on PMN activation. Results were compared against C57BL/6 age and sex-matched controls. RESULTS: Relative to wild-type controls, hSOD3 heterozygous mice exhibited a reduction in lung inflammation, blood-brain barrier damage, and post-ischemic neuronal injury within the hippocampus and cortex. PMNs harvested from hSOD3 mice were also resistant to LPS priming, slower-moving, and less toxic to primary neuronal cultures. CONCLUSIONS: Constitutive, focal expression of hSOD3 is neuroprotective in a model of global cerebral ischemia-reperfusion injury. The underlying mechanism of SOD3-dependent protection is attributable in part to effects on the activation state and toxic potential of circulating neutrophils. These results implicate lung-brain coupling as a determinant of cerebral ischemia-reperfusion injury and highlight post-stroke lung inflammation as a potential therapeutic target in acute ischemic cerebrovascular injuries.


Asunto(s)
Células Epiteliales Alveolares/enzimología , Isquemia Encefálica/enzimología , Encéfalo/metabolismo , Neuronas/metabolismo , Activación Neutrófila , Neutrófilos/metabolismo , Neumonía/prevención & control , Daño por Reperfusión/prevención & control , Superóxido Dismutasa/metabolismo , Células Epiteliales Alveolares/patología , Animales , Encéfalo/patología , Isquemia Encefálica/genética , Isquemia Encefálica/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/patología , Neutrófilos/inmunología , Neumonía/enzimología , Neumonía/genética , Neumonía/inmunología , Daño por Reperfusión/enzimología , Daño por Reperfusión/genética , Daño por Reperfusión/inmunología , Transducción de Señal , Superóxido Dismutasa/genética
8.
Am J Respir Cell Mol Biol ; 61(4): 481-491, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30917006

RESUMEN

The discovery of mutant tyrosine kinases as oncogenic drivers of lung adenocarcinomas has changed the basic understanding of lung cancer development and therapy. Yet, expressed kinases (kinome) in lung cancer progenitor cells, as well as whether kinase expression and the overall kinome changes or is reprogrammed upon transformation, is incompletely understood. We hypothesized that the kinome differs between lung cancer progenitor cells, alveolar type II cells (ATII), and basal cells (BC) and that their respective kinomes undergo distinct lineage-specific reprogramming to adenocarcinomas and squamous cell carcinomas upon transformation. We performed RNA sequencing on freshly isolated human ATII, BC, and lung cancer cell lines to define the kinome in nontransformed cells and transformed cells. Our studies identified a unique kinome for ATII and BC and changes in their kinome upon transformation to their respective carcinomas.


Asunto(s)
Células Madre Adultas/enzimología , Células Epiteliales Alveolares/enzimología , Transformación Celular Neoplásica , Neoplasias Pulmonares/enzimología , Pulmón/enzimología , Proteínas de Neoplasias/análisis , Proteínas Tirosina Quinasas/análisis , Adenocarcinoma/enzimología , Adenocarcinoma/genética , Animales , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/genética , Linaje de la Célula , Células Cultivadas , Inducción Enzimática , Humanos , Pulmón/citología , Neoplasias Pulmonares/genética , Ratones , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/enzimología , Proteínas Tirosina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/genética , ARN Mensajero/análisis , ARN Neoplásico/análisis , Transcriptoma
9.
Biochem Biophys Res Commun ; 498(4): 960-966, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29548827

RESUMEN

Infection with influenza A virus (IAV) A/WSN/1933 (H1N1) causes oxidative stress and severe lung injury. We have demonstrated that the generation of reactive oxygen species (ROS) during IAV infection is tightly regulated by superoxide dismutase 1 (SOD1) and correlated with viral replication in alveolar epithelial cells. However, the molecular mechanism underlying SOD1 reduction during IAV infection is uncertain. Here we demonstrate that the autophagy pathway is activated by IAV infection and involved in enhanced ROS generation in the early phase of infection. We observed that IAV infection induced autophagic vacuolation, leading to autophagic degradation of cellular proteins, including the protease sensitive antioxidant SOD1. Silencing of the microtubule-associated protein 1A/1B-light chain 3 (LC3) gene in A549 cells supported the critical role of autophagy in the ROS increase. The decrease in viral titer and viral polymerase activity caused by LC3 silencing or the autophagy inhibitor clearly evidenced the involvement of autophagy in the control of ROS generation and viral infectivity. Therefore, we concluded that early stage IAV infection induces autophagic degradation of antioxidant enzyme SOD1, thereby contributing to increased ROS generation and viral infectivity in alveolar epithelial cells.


Asunto(s)
Células Epiteliales Alveolares/enzimología , Células Epiteliales Alveolares/virología , Autofagia/fisiología , Virus de la Influenza A/patogenicidad , Gripe Humana/etiología , Superóxido Dismutasa-1/metabolismo , Células A549 , Regulación hacia Abajo , Silenciador del Gen , Interacciones Huésped-Patógeno , Humanos , Proteínas Asociadas a Microtúbulos/genética , Especies Reactivas de Oxígeno
10.
Am J Respir Cell Mol Biol ; 54(3): 319-30, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26203800

RESUMEN

Type 2 alveolar epithelial cells (AEC2) are regarded as the progenitor population of the alveolus responsible for injury repair and homeostatic maintenance. Depletion of this population is hypothesized to underlie various lung pathologies. Current models of lung injury rely on either uncontrolled, nonspecific destruction of alveolar epithelia or on targeted, nontitratable levels of fixed AEC2 ablation. We hypothesized that discrete levels of AEC2 ablation would trigger stereotypical and informative patterns of repair. To this end, we created a transgenic mouse model in which the surfactant protein-C promoter drives expression of a mutant SR39TK herpes simplex virus-1 thymidine kinase specifically in AEC2. Because of the sensitivity of SR39TK, low doses of ganciclovir can be administered to these animals to induce dose-dependent AEC2 depletion ranging from mild (50%) to lethal (82%) levels. We demonstrate that specific levels of AEC2 depletion cause altered expression patterns of apoptosis and repair proteins in surviving AEC2 as well as distinct changes in distal lung morphology, pulmonary function, collagen deposition, and expression of remodeling proteins in whole lung that persist for up to 60 days. We believe SPCTK mice demonstrate the utility of cell-specific expression of the SR39TK transgene for exerting fine control of target cell depletion. Our data demonstrate, for the first time, that specific levels of type 2 alveolar epithelial cell depletion produce characteristic injury repair outcomes. Most importantly, use of these mice will contribute to a better understanding of the role of AEC2 in the initiation of, and response to, lung injury.


Asunto(s)
Células Epiteliales Alveolares/patología , Lesión Pulmonar/patología , Fibrosis Pulmonar/patología , Regeneración , Células Epiteliales Alveolares/enzimología , Animales , Apoptosis , Proliferación Celular , Forma de la Célula , Células Cultivadas , Colágeno/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ganciclovir/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Predisposición Genética a la Enfermedad , Humanos , Hiperoxia/complicaciones , Lesión Pulmonar/genética , Lesión Pulmonar/metabolismo , Lesión Pulmonar/fisiopatología , Ratones Transgénicos , Fenotipo , Regiones Promotoras Genéticas , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/fisiopatología , Proteína C Asociada a Surfactante Pulmonar/genética , Simplexvirus/enzimología , Simplexvirus/genética , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Factores de Tiempo , Proteínas Virales/genética , Proteínas Virales/metabolismo
11.
Cell Biol Toxicol ; 32(5): 437-49, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27344565

RESUMEN

Calcium signaling participates in the regulation of numberless cellular functions including cell cycle progression and cellular migration, important processes for cancer expansion. Cancer cell growth, migration, and invasion are typically supported by PI3K/Akt activation, while a hypoxic environment is critical in cancer development. Accordingly, in the present study, we aimed at investigating whether perturbations in calcium homeostasis induce alterations of HIF-1α and activate Akt levels in epithelial A549 and A431 cells. Survival was drastically reduced in the presence of calcium chelator BAPTA-AM and thapsigargin, a SERCA inhibitor inducing store-operated calcium entry, to a lesser extent. Calcium chelation provoked a transient but strong upregulation of HIF-1α protein levels and accumulation in the nucleus, whereas in the presence of thapsigargin, HIF-1α levels were rapidly abolished before reaching and exceeding control levels. Despite cell death, calcium chelation merely inhibited Akt, which was significantly activated in the presence of thapsigargin. Moreover, when store-operated calcium entry was simulated by reintroducing calcium ions in cell suspensions, Akt was rapidly activated in the absence of any growth factor. These data further underscore the growing importance of calcium entry and directly link this elementary event of calcium homeostasis to the Akt pathway, which is commonly deregulated in cancer.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Calcio/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células A549 , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/enzimología , Señalización del Calcio , Línea Celular Tumoral , Quelantes/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Tapsigargina/farmacología , Activación Transcripcional
12.
Am J Respir Cell Mol Biol ; 53(6): 769-81, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25906418

RESUMEN

Chronic obstructive pulmonary disease (COPD) is characterized by an irreversible loss of lung function and is one of the most prevalent and severe diseases worldwide. A major feature of COPD is emphysema, which is the progressive loss of alveolar tissue. Coactivator-associated arginine methyltransferase-1 (CARM1) regulates histone methylation and the transcription of genes involved in senescence, proliferation, and differentiation. Complete loss of CARM1 leads to disrupted differentiation and maturation of alveolar epithelial type II (ATII) cells. We thus hypothesized that CARM1 regulates the development and progression of emphysema. To address this, we investigated the contribution of CARM1 to alveolar rarefication using the mouse model of elastase-induced emphysema in vivo and small interfering (si)RNA-mediated knockdown in ATII-like LA4 cells in vitro. We demonstrate that emphysema progression in vivo is associated with a time-dependent down-regulation of CARM1. Importantly, elastase-treated CARM1 haploinsufficient mice show significantly increased airspace enlargement (52.5 ± 9.6 µm versus 38.8 ± 5.5 µm; P < 0.01) and lung compliance (2.8 ± 0.32 µl/cm H2O versus 2.4 ± 0.4 µl/cm H2O; P < 0.04) compared with controls. The knockdown of CARM1 in LA4 cells led to decreased sirtuin 1 expression (0.034 ± 0.003 versus 0.022 ± 0.001; P < 0.05) but increased expression of p16 (0.27 ± 0.013 versus 0.31 ± 0.010; P < 0.5) and p21 (0.81 ± 0.088 versus 1.28 ± 0.063; P < 0.01) and higher ß-galactosidase-positive senescent cells (50.57 ± 7.36% versus 2.21 ± 0.34%; P < 0.001) compared with scrambled siRNA. We further demonstrated that CARM1 haploinsufficiency impairs transdifferentiation and wound healing (32.18 ± 0.9512% versus 8.769 ± 1.967%; P < 0.001) of alveolar epithelial cells. Overall, these results reveal a novel function of CARM1 in regulating emphysema development and premature lung aging via alveolar senescence as well as impaired regeneration, repair, and differentiation of ATII cells.


Asunto(s)
Células Epiteliales Alveolares/enzimología , Proteína-Arginina N-Metiltransferasas/fisiología , Enfisema Pulmonar/enzimología , Animales , Diferenciación Celular , Línea Celular , Senescencia Celular , Femenino , Predisposición Genética a la Enfermedad , Haploinsuficiencia , Ratones Endogámicos C57BL , Elastasa Pancreática , Enfisema Pulmonar/inducido químicamente
13.
Am J Respir Cell Mol Biol ; 52(1): 25-36, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24918270

RESUMEN

Asbestos causes asbestosis and malignancies by mechanisms that are not fully established. Alveolar epithelial cell (AEC) injury and repair are crucial determinants of the fibrogenic potential of noxious agents such as asbestos. We previously showed that mitochondrial reactive oxygen species mediate asbestos-induced AEC intrinsic apoptosis and that mitochondrial human 8-oxoguanine-DNA glycosylase 1 (OGG1), a DNA repair enzyme, prevents oxidant-induced AEC apoptosis. We reasoned that OGG1 deficiency augments asbestos-induced pulmonary fibrosis. Compared with intratracheal instillation of PBS (50 µl) or titanium dioxide (100 µg/50 µl), crocidolite or Libby amphibole asbestos (100 µg/50 µl) each augmented pulmonary fibrosis in wild-type C57BL/6J (WT) mice after 3 weeks as assessed by histology, fibrosis score, lung collagen via Sircol, and type 1 collagen expression; these effects persisted at 2 months. Compared with WT mice, Ogg1 homozygous knockout (Ogg1(-/-)) mice exhibit increased pulmonary fibrosis after crocidolite exposure and apoptosis in cells at the bronchoalveolar duct junctions as assessed via cleaved caspase-3 immunostaining. AEC involvement was verified by colocalization studies using surfactant protein C. Asbestos increased endoplasmic reticulum stress in the lungs of WT and Ogg1(-/-) mice. Compared with WT, alveolar type 2 cells isolated from Ogg1(-/-) mice have increased mtDNA damage, reduced mitochondrial aconitase expression, and increased P53 and cleaved caspase-9 expression, and these changes were enhanced 3 weeks after crocidolite exposure. These findings suggest an important role for AEC mtDNA integrity maintained by OGG1 in the pathogenesis of pulmonary fibrosis that may represent a novel therapeutic target.


Asunto(s)
Células Epiteliales Alveolares/enzimología , Asbesto Crocidolita/toxicidad , ADN Glicosilasas/metabolismo , Fibrosis Pulmonar/enzimología , Células Epiteliales Alveolares/patología , Animales , Daño del ADN/genética , ADN Glicosilasas/genética , ADN Glicosilasas/inmunología , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Humanos , Ratones , Ratones Noqueados , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Factores de Tiempo
14.
Am J Respir Cell Mol Biol ; 61(4): 537-540, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31573336
15.
Respir Res ; 15: 106, 2014 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-25186164

RESUMEN

BACKGROUND: Chronic pulmonary obstructive disease (COPD) has become the fourth leading cause of death worldwide. Cigarette smoking induces neutrophil elastase (NE) and contributes to COPD, but the detailed mechanisms involved are not fully established. In an animal model of pulmonary emphysema, there are increased expressions of placenta growth factor (PlGF) and lung epithelial (LE) cell apoptosis. This study hypothesized that excessive NE may up-regulate PlGF and that PlGF-induced LE apoptosis mediates the pathogenesis of pulmonary emphysema. METHODS: Human bronchial epithelial cells, BEAS-2B, and primary mouse type II alveolar epithelial cells were treated with NE. The PlGF promoter activity was examined by luciferase activity assay, while PlGF expression and secretion were evaluated by RT-PCR, Western blotting, and ELISA. Both cell lines were treated with PlGF to evaluate its effects and the downstream signaling pathways leading to LE cell apoptosis. PlGF knockout and wild-type mice were instilled with NE to determine the roles of PlGF and its downstream molecules in NE-promoted mice pulmonary apoptosis and emphysema phenotype. RESULTS: The transcriptional factor, early growth response gene-1, was involved in the NE-promoted PlGF promoter activity, and the expression and secretion of PlGF mRNA and protein in LE cells. PlGF-induced LE cell apoptosis and NE-induced mice pulmonary apoptosis and emphysema were mediated by the downstream c-Jun N-terminal kinase (JNK) and protein kinase C (PKC)δ signaling pathways. CONCLUSION: The NE-PlGF-JNK/PKCδ pathway contributes to the pathogenesis of LE cell apoptosis and emphysema. PlGF and its downstream signaling molecules may be potential therapeutic targets for COPD.


Asunto(s)
Células Epiteliales Alveolares/enzimología , Apoptosis , Elastasa de Leucocito/metabolismo , Proteínas Gestacionales/metabolismo , Enfisema Pulmonar/enzimología , Transducción de Señal , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/inmunología , Células Epiteliales Alveolares/patología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Elastasa de Leucocito/farmacología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Crecimiento Placentario , Proteínas Gestacionales/genética , Proteínas Gestacionales/farmacología , Regiones Promotoras Genéticas , Proteína Quinasa C-delta/metabolismo , Enfisema Pulmonar/genética , Enfisema Pulmonar/inmunología , Enfisema Pulmonar/patología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección , Regulación hacia Arriba
16.
Lung ; 192(4): 467-72, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24792232

RESUMEN

BACKGROUND: An imbalance between proteolytic enzymes and their inhibitors is thought to be involved in the pathogenesis of chronic obstructive pulmonary disease. Matrix metalloproteinase-1, also known as interstitial collagenase, has been implicated as a potentially important proteinase in the genesis of chronic obstructive pulmonary disease and, more specifically, emphysema. METHODS: We performed quantitative immunohistochemical assessment of matrix metalloproteinase-1 expression in the resected lung of 20 smokers/ex-smokers who had varying severity of airflow obstruction and emphysema and compared this with the lungs of 5 nonsmokers. Emphysema was measured using a morphometric measure of the lungs' surface area/volume ratio and with qualitative and quantitative computed tomography (CT) measures of emphysema. RESULTS: There were significantly more matrix metalloproteinase-1-expressing alveolar macrophages and type II pneumocytes as well as a greater percentage of small airways that stained positively for matrix metalloproteinase-1 in the lungs of smokers than in those of nonsmokers (p < 0.0001, p < 0.0001, and p = 0.0003, respectively). The extent of staining of type II pneumocytes and airways for matrix metalloproteinase-1 was significantly related to the extent of smoking (p = 0.012 and p = 0.013, respectively). In addition, the extent of matrix metalloproteinase-1 staining of alveolar macrophages was related to the lung surface area/volume ratio and to qualitative estimates of emphysema on CT. CONCLUSION: These findings suggest that cigarette smoking increases expression of matrix metalloproteinase-1 in alveolar macrophages as well as in alveolar and small airway epithelial cells. Smokers who develop emphysema have increased alveolar macrophage expression of matrix metalloproteinase-1.


Asunto(s)
Células Epiteliales Alveolares/enzimología , Pulmón/enzimología , Macrófagos Alveolares/enzimología , Metaloproteinasa 1 de la Matriz/análisis , Enfisema Pulmonar/enzimología , Fumar/metabolismo , Adulto , Anciano , Estudios de Casos y Controles , Femenino , Humanos , Inmunohistoquímica , Pulmón/diagnóstico por imagen , Pulmón/fisiopatología , Masculino , Persona de Mediana Edad , Enfisema Pulmonar/diagnóstico por imagen , Enfisema Pulmonar/etiología , Enfisema Pulmonar/fisiopatología , Pruebas de Función Respiratoria , Índice de Severidad de la Enfermedad , Fumar/efectos adversos , Fumar/fisiopatología , Tomografía Computarizada por Rayos X , Regulación hacia Arriba
17.
Am J Respir Cell Mol Biol ; 48(5): 626-34, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23349050

RESUMEN

Elevated concentrations of CO2 (hypercapnia) lead to alveolar epithelial dysfunction by promoting Na,K-ATPase endocytosis. In the present report, we investigated whether the CO2/HCO3(-) activated soluble adenylyl cyclase (sAC) regulates this process. We found that hypercapnia increased the production of cyclic adenosine monophosphate (cAMP) and stimulated protein kinase A (PKA) activity via sAC, which was necessary for Na,K-ATPase endocytosis. During hypercapnia, cAMP was mainly produced in specific microdomains in the proximity of the plasma membrane, leading to PKA Type Iα activation. In alveolar epithelial cells exposed to high CO2 concentrations, PKA Type Iα regulated the time-dependent phosphorylation of the actin cytoskeleton component α-adducin at serine 726. Cells expressing small hairpin RNA for PKAc, dominant-negative PKA Type Iα, small interfering RNA for α-adducin, and α-adducin with serine 726 mutated to alanine prevented Na,K-ATPase endocytosis. In conclusion, we provide evidence for a new mechanism by which hypercapnia via sAC, cAMP, PKA Type Iα, and α-adducin regulates Na,K-ATPase endocytosis in alveolar epithelial cells.


Asunto(s)
Células Epiteliales Alveolares/enzimología , Dióxido de Carbono/farmacología , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/fisiología , Endocitosis , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Adenilil Ciclasas/metabolismo , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/fisiología , Animales , Proteínas de Unión a Calmodulina/metabolismo , Línea Celular Tumoral , Membrana Celular/enzimología , AMP Cíclico/metabolismo , Humanos , Hipercapnia/enzimología , Fosforilación , Proteína Quinasa C/metabolismo , Procesamiento Proteico-Postraduccional , Ratas , Ratas Sprague-Dawley , Sistemas de Mensajero Secundario
18.
Biochim Biophys Acta ; 1823(10): 1796-806, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22713544

RESUMEN

Lung surfactant secretion involves lamellar body docking and fusion with the plasma membrane in alveolar type II cells. Annexin A7 (A7) is postulated to play a role in membrane fusion during exocytosis. Our recent studies demonstrated increased co-localization of A7 with ABCA3 in lamellar bodies in type II cells stimulated with established secretagogues of lung surfactant. In this study, we investigated in vivo and in vitro interactions of A7 with the t-SNARE protein, SNAP23. Immuno-fluorescence studies showed time-dependent increases in co-localization of A7 with SNAP23 in PMA- and in A23187-stimulated cells. PMA and A23187 also caused a time-dependent increase in co-localization of ABCA3 with SNAP23. The relocation of A7 to SNAP23 domains was inhibited in the presence of PKC inhibitor, similar to that previously reported for co-localization of A7 with ABCA3. The interaction of A7 and SNAP23 was confirmed by affinity binding and by in vitro interaction of recombinant A7 and SNAP23 proteins. The in vitro binding of recombinant A7 (rA7) to GST-SNAP23 fusion protein was calcium-dependent. Phosphorylation of rA7 with PKC increased its in vitro binding to SNAP23 suggesting that a similar mechanism may operate during A7 relocation to t-SNARE domains. Thus, our studies demonstrate that annexin A7 may function in co-ordination with SNARE proteins and that protein kinase activation may be required for annexin A7 trafficking to the interacting membranes (lamellar bodies and plasma membrane) to facilitate membrane fusion during surfactant secretion.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Anexina A7/metabolismo , Calcio/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/enzimología , Animales , Anexina A7/inmunología , Anticuerpos/inmunología , Calcimicina/farmacología , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Proteínas SNARE/química , Proteínas SNARE/metabolismo , Acetato de Tetradecanoilforbol/farmacología
19.
Glycobiology ; 23(6): 709-19, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23418199

RESUMEN

Previously, we have shown that heparan sulfate (HS) 6-O-endosulfatase 1 (Sulf1) is a transforming growth factor-ß1 (TGF-ß1)-responsive gene in normal human lung fibroblasts and functions as a negative feedback regulator of TGF-ß1 and that TGF-ß1 induces the expression of Sulf1 as well as that of the closely related Sulf2 in a murine model of pulmonary fibrosis. In this study, we focused on the role of Sulf2 in modulating TGF-ß1 function and the development of pulmonary fibrosis. We found that Sulf2 mRNA was overexpressed in lung samples from human patients with idiopathic pulmonary fibrosis (IPF), and Sulf2 protein was specifically localized to the hyperplastic type II alveolar epithelial cells (AECs). In vitro, TGF-ß1 induced the expression of Sulf2 with accompanied HS 6-O-desulfation in A549 cells, adenocarcinoma cells derived from the type II alveolar epithelium. Using small interference RNA to block Sulf2 expression, we observed a biphasic TGF-ß1 response with early enhanced Smad activation, but eventually reduced TGF-ß1 target gene expression in Sulf2 knockdown A549 cells compared with the control cells. To study the role of Sulf2 in normal type II AECs, we isolated primary type II cells from wild-type and Sulf2 knockout mice. We observed enhanced Smad activation as well as enhanced TGF-ß1 target gene expression in Sulf2 knockout type II AECs compared with wild-type type II AECs. In conclusion, Sulf2 is overexpressed in IPF and may play a role in regulating TGF-ß1 signaling in type II AECs.


Asunto(s)
Fibrosis Pulmonar Idiopática/enzimología , Sulfotransferasas/metabolismo , Anciano , Células Epiteliales Alveolares/enzimología , Animales , Antibióticos Antineoplásicos/farmacología , Bleomicina/farmacología , Línea Celular Tumoral , Inducción Enzimática , Femenino , Expresión Génica/efectos de los fármacos , Heparitina Sulfato/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Cultivo Primario de Células , Transducción de Señal , Proteínas Smad/metabolismo , Sulfatasas , Sulfotransferasas/genética , Factor de Crecimiento Transformador beta1/fisiología
20.
J Toxicol Environ Health A ; 76(24): 1317-32, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24283474

RESUMEN

Carbon black nanoparticle (CBNP) applications in high doses have been shown to be harmful to the lung. It is postulated that even small, environmentally relevant concentrations induce changes on lung homeostasis. The present study determined the impact of low-dose single and multiple CBNP (Printex 90) applications on mouse alveolar cell metabolism, especially inflammatory and oxidative stress parameters. Nanoparticles were administered to mice by a single or 8 oropharyngeal aspirations at wk 1, 2, 3, 5, 7, 9, 11, and 12 using 7 µg Printex 90, 7 µg DQ12 quartz (positive control), with water vehicle and saline as negative controls. After 2 d or 3 mo lung function was analyzed. Further lung histology, bronchoalveolar lavage fluid (BALF) parameters, and mRNA expression of cytokines and antioxidants enzymes in type II pneumocytes were measured on d 3 or after 3 mo. Single low-dose Printex 90 application induced no marked alterations in lung functions or BALF phospholipid levels but significant decrease in superoxide dismutase 2 and numerically elevated glutathione peroxidase 3 mRNA expression levels in type II pneumocytes. Multiple CBNP applications produced reduced lung function, collagen accumulation, elevated phospholipid levels in BALF, and a massive infiltration of macrophages. Type II pneumocyte mRNA expression of antioxidative enzymes remained unchanged throughout the subchronic experiment, but showed a significant decrease in interleukin (IL)-6Rα mRNA expression. This study demonstrates that an environmentally relevant CBNP concentration induced an acute inflammatory response, an effect that is exacerbated throughout the subchronic duration.


Asunto(s)
Exposición por Inhalación , Pulmón/efectos de los fármacos , Nanopartículas/toxicidad , Alveolos Pulmonares/efectos de los fármacos , Hollín/toxicidad , Células Epiteliales Alveolares/citología , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/enzimología , Células Epiteliales Alveolares/metabolismo , Animales , Antioxidantes/metabolismo , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Citocinas/genética , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Pulmón/inmunología , Pulmón/fisiopatología , Ratones , Ratones Endogámicos BALB C , Pletismografía Total , Reacción en Cadena de la Polimerasa , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/fisiopatología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Organismos Libres de Patógenos Específicos , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA