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












Base de datos
Intervalo de año de publicación
2.
Physiol Rep ; 9(9): e14854, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33991451

RESUMEN

SARS-CoV-2 uptake by lung epithelial cells is a critical step in the pathogenesis of COVID-19. Viral entry is dependent on the binding of the viral spike protein to the angiotensin converting enzyme II protein (ACE2) on the host cell surface, followed by proteolytic cleavage by a host serine protease such as TMPRSS2. Infection of alveolar epithelial cells (AEC) in the distal lung is a key feature in progression to the acute respiratory distress syndrome (ARDS). We hypothesized that AEC expression of ACE2 is induced by hypoxia. In a murine model of hypoxic stress (12% FiO2), the total lung Ace2 mRNA and protein expression was significantly increased after 24 hours in hypoxia compared to normoxia (21% FiO2). In experiments with primary murine type II AEC, we found that exposure to hypoxia either in vivo (prior to isolation) or in vitro resulted in greatly increased AEC expression of both Ace2 (mRNA and protein) and of Tmprss2. However, when isolated type II AEC were maintained in culture over 5 days, with loss of type II cell characteristics and induction of type I cell features, Ace2 expression was greatly reduced, suggesting that this expression was a feature of only this subset of AEC. Finally, in primary human small airway epithelial cells (SAEC), ACE2 mRNA and protein expression were also induced by hypoxia, as was binding to purified spike protein. Hypoxia-induced increase in ACE2 expression in type II AEC may provide an explanation of the extended temporal course of human patients who develop ARDS in COVID-19.


Asunto(s)
Lesión Pulmonar Aguda/enzimología , Células Epiteliales Alveolares/enzimología , Enzima Convertidora de Angiotensina 2/biosíntesis , COVID-19/enzimología , Regulación Enzimológica de la Expresión Génica , Hipoxia/enzimología , Lesión Pulmonar Aguda/genética , Enzima Convertidora de Angiotensina 2/genética , Animales , COVID-19/genética , Células Cultivadas , Femenino , Humanos , Hipoxia/genética , Masculino , Ratones , Ratones Endogámicos C57BL
3.
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
4.
Life Sci ; 278: 119032, 2021 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-33450260

RESUMEN

Silica (SiO2) nanoparticles (NPs) were synthesized by laser ablation method and were characterized by TEM and DLS techniques. Afterwards, their inhibition activity against carbonic anhydrase (CA) isoforms (CA I and CA II) was explored by experimental and theoretical analysis. Also, the protective effect of SiO2 NPs against H2O2-induced oxidative stress in alveolar epithelial cells (A549) were assessed by measurement of MTT, ROS level, CAT and SOD activity and GSH content. Finally, the NPs were screened for their antimicrobial activity using the MICs method against the Klebsiella pneumoniae. The result showed that the synthesized NPs have a size of around 40 nm. The inhibition activity by comparing IC50 values with acetazolamide as a positive control revealed that SiO2 NPs in comparison with acetazolamide served as potent inhibitors against CA isoforms which was also confirmed by docking studies. The cellular assays indicated that the SiO2 NPs with a concentration of 20 µg/mL stimulated a significant antioxidant activity against H2O2-induced oxidative cell damage through activation of CAT and SOD, an increase in the GSH content and reducing the level of ROS. The synthesize NPs also showed a good inhibition effect against Klebsiella pneumoniae as compared to Sulfamethoxazole as a positive control. In conclusion, this data may provide some useful information on the development of some platforms for pneumonia treatment and management.


Asunto(s)
Células Epiteliales Alveolares/efectos de los fármacos , Antibacterianos/farmacología , Antioxidantes/farmacología , Inhibidores de Anhidrasa Carbónica/farmacología , Anhidrasas Carbónicas/química , Infecciones por Klebsiella/tratamiento farmacológico , Nanopartículas/administración & dosificación , Dióxido de Silicio/química , Células A549 , Células Epiteliales Alveolares/enzimología , Células Epiteliales Alveolares/microbiología , Humanos , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/efectos de los fármacos , Terapia por Láser , Nanopartículas/química , Nanopartículas/efectos de la radiación
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.
J Clin Invest ; 131(1)2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33180746

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) novel coronavirus 2019 (COVID-19) global pandemic has led to millions of cases and hundreds of thousands of deaths. While older adults appear at high risk for severe disease, hospitalizations and deaths due to SARS-CoV-2 among children have been relatively rare. Integrating single-cell RNA sequencing (scRNA-seq) of developing mouse lung with temporally resolved immunofluorescence in mouse and human lung tissue, we found that expression of SARS-CoV-2 Spike protein primer TMPRSS2 was highest in ciliated cells and type I alveolar epithelial cells (AT1), and TMPRSS2 expression increased with aging in mice and humans. Analysis of autopsy tissue from fatal COVID-19 cases detected SARS-CoV-2 RNA most frequently in ciliated and secretory cells in airway epithelium and AT1 cells in peripheral lung. SARS-CoV-2 RNA was highly colocalized in cells expressing TMPRSS2. Together, these data demonstrate the cellular spectrum infected by SARS-CoV-2 in lung epithelium and suggest that developmental regulation of TMPRSS2 may underlie the relative protection of infants and children from severe respiratory illness.


Asunto(s)
Células Epiteliales Alveolares/enzimología , COVID-19/enzimología , COVID-19/metabolismo , Regulación Enzimológica de la Expresión Génica , SARS-CoV-2/metabolismo , Serina Endopeptidasas/biosíntesis , Adulto , Envejecimiento , Células Epiteliales Alveolares/patología , Células Epiteliales Alveolares/virología , Animales , COVID-19/patología , Preescolar , Modelos Animales de Enfermedad , Femenino , Humanos , Lactante , Masculino , Ratones
7.
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
8.
Cell Stem Cell ; 27(6): 890-904.e8, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33128895

RESUMEN

Coronavirus infection causes diffuse alveolar damage leading to acute respiratory distress syndrome. The absence of ex vivo models of human alveolar epithelium is hindering an understanding of coronavirus disease 2019 (COVID-19) pathogenesis. Here, we report a feeder-free, scalable, chemically defined, and modular alveolosphere culture system for the propagation and differentiation of human alveolar type 2 cells/pneumocytes derived from primary lung tissue. Cultured pneumocytes express the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) receptor angiotensin-converting enzyme receptor type-2 (ACE2) and can be infected with virus. Transcriptome and histological analysis of infected alveolospheres mirror features of COVID-19 lungs, including emergence of interferon (IFN)-mediated inflammatory responses, loss of surfactant proteins, and apoptosis. Treatment of alveolospheres with IFNs recapitulates features of virus infection, including cell death. In contrast, alveolospheres pretreated with low-dose IFNs show a reduction in viral replication, suggesting the prophylactic effectiveness of IFNs against SARS-CoV-2. Human stem cell-based alveolospheres, thus, provide novel insights into COVID-19 pathogenesis and can serve as a model for understanding human respiratory diseases.


Asunto(s)
Células Madre Adultas/virología , Células Epiteliales Alveolares/efectos de los fármacos , Tratamiento Farmacológico de COVID-19 , Interferones/farmacología , SARS-CoV-2/inmunología , Adulto , Células Madre Adultas/efectos de los fármacos , Células Madre Adultas/enzimología , Anciano , Anciano de 80 o más Años , Células Epiteliales Alveolares/enzimología , Células Epiteliales Alveolares/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/fisiopatología , Técnicas de Cultivo de Célula , Diferenciación Celular , Femenino , Humanos , Inflamación , Masculino , Ratones , Receptores de Coronavirus/metabolismo , Transcriptoma , Replicación Viral
9.
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
11.
Virchows Arch ; 477(4): 557-563, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32270298

RESUMEN

Surfactant protein B (SP-B) is a key component of pulmonary surfactant. SP-B is processed to a mature, surface-active protein from a pro-peptide by two distinct cleavage events in its N-terminal and C-terminal regions. Napsin A, a protease expressed in type II pneumocytes, is responsible for the N-terminal cleavage event. Here, for the first time, we have evaluated the expression of Napsin A in normal fetal lungs at different gestational ages and in lungs from fetuses and neonates with congenital and acquired pathological pulmonary conditions. Lung samples were collected from fetal and neonatal autopsies at the Department of Medicine and Surgery's Pathology Unit of Parma University (Italy). Immunohistochemical analysis was performed using a primary anti-Napsin A (clone IP64 clone) monoclonal antibody. A section of lung adenocarcinoma was used as an external positive control. Napsin A was expressed early in normal fetal lungs throughout the epithelium of the distal pseudoglandular tracts. In fetuses at 30 weeks of gestation and term newborns, Napsin A was already expressed only in isolated cells within the alveolar epithelium, similar to adult subjects. Furthermore, increased expression of Napsin A compared with a control group was observed in lung tissue from fetuses and a newborn with pathological conditions (inflammatory diseases and pulmonary hypoplasia). In conclusion, this study demonstrates that Napsin A is produced early in fetal life, and that its production is increased in many diseases, presumably in an effort to remedy functional pulmonary failure.


Asunto(s)
Células Epiteliales Alveolares/enzimología , Ácido Aspártico Endopeptidasas/análisis , Inmunohistoquímica , Enfermedades Pulmonares/enzimología , Pulmón/enzimología , Autopsia , Biomarcadores/análisis , Edad Gestacional , Humanos , Recién Nacido , Pulmón/anomalías , Enfermedades Pulmonares/congénito , Enfermedades Pulmonares/mortalidad , Valor Predictivo de las Pruebas , Regulación hacia Arriba
12.
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
13.
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
14.
Sci Rep ; 10(1): 419, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31942012

RESUMEN

Mechanical ventilation induces lung injury by damaging alveolar epithelial cells (AECs), but the pathogenesis remains unknown. Focal adhesion kinase (FAK) is a cytoplasmic protein tyrosine kinase that is involved in cell growth and intracellular signal transduction pathways. This study explored the potential role of FAK in AECs during lung injury induced by mechanical ventilation. High-volume mechanical ventilation (HMV) was used to create a mouse lung injury model, which was validated by analysis of lung weight, bronchoalveolar lavage fluid and histological investigation. The expression of FAK and Akt in AECs were evaluated. In addition, recombinant FAK was administered to mice via the tail vein, and then the extent of lung injury was assessed. Mouse AECs were cultured in vitro, and FAK expression in cells under stretch was investigated. The effects of FAK on cell proliferation, migration and apoptosis were investigated. The results showed that HMV decreased FAK expression in AECs of mice, while FAK supplementation attenuated lung injury, reduced protein levels/cell counts in the bronchoalveolar lavage fluid and decreased histological lung injury and oedema. The protective effect of FAK promoted AEC proliferation and migration and prevented cells from undergoing apoptosis, which restored the integrity of the alveoli through Akt pathway. Therefore, the decrease in FAK expression by HMV is essential for injury to epithelial cells and the disruption of alveolar integrity. FAK supplementation can reduce AEC injury associated with HMV.


Asunto(s)
Células Epiteliales Alveolares/patología , Modelos Animales de Enfermedad , Quinasa 1 de Adhesión Focal/metabolismo , Lesión Pulmonar/prevención & control , Ventiladores Mecánicos/efectos adversos , Cicatrización de Heridas , Células Epiteliales Alveolares/enzimología , Animales , Apoptosis , Proliferación Celular , Células Cultivadas , Quinasa 1 de Adhesión Focal/genética , Lesión Pulmonar/etiología , Lesión Pulmonar/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal
15.
Am J Respir Cell Mol Biol ; 61(4): 537-540, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31573336
16.
Sci Rep ; 9(1): 12509, 2019 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-31467330

RESUMEN

Proteasomes are a critical component of quality control that regulate turnover of short-lived, unfolded, and misfolded proteins. Proteasome activity has been therapeutically targeted and considered as a treatment option for several chronic lung disorders including pulmonary fibrosis. Although pharmacologic inhibition of proteasome activity effectively prevents the transformation of fibroblasts to myofibroblasts, the effect on alveolar type 2 (AT2) epithelial cells is not clear. To address this knowledge gap, we generated a genetic model in which a proteasome subunit, RPT3, which promotes assembly of active 26S proteasome, was conditionally deleted in AT2 cells of mice. Partial deletion of RPT3 resulted in 26S proteasome dysfunction, leading to augmented cell stress and cell death. Acute loss of AT2 cells resulted in depletion of alveolar surfactant, disruption of the alveolar epithelial barrier and, ultimately, lethal acute respiratory distress syndrome (ARDS). This study underscores importance of proteasome function in maintenance of AT2 cell homeostasis and supports the need to further investigate the role of proteasome dysfunction in ARDS pathogenesis.


Asunto(s)
Células Epiteliales Alveolares/enzimología , Complejo de la Endopetidasa Proteasomal/metabolismo , Síndrome de Dificultad Respiratoria/enzimología , Células Epiteliales Alveolares/citología , Animales , Muerte Celular , Femenino , Fibroblastos/citología , Fibroblastos/enzimología , Eliminación de Gen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Miofibroblastos/citología , Miofibroblastos/enzimología , Complejo de la Endopetidasa Proteasomal/genética , Síndrome de Dificultad Respiratoria/genética , Síndrome de Dificultad Respiratoria/fisiopatología
17.
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
18.
Int J Mol Med ; 44(1): 273-280, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31115510

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease characterized by progressive lung scarring and excessive extracellular matrix depositon. When stimulated, alveolar epithelial cells (AECs) are aberrantly activated, the expression of profibrotic molecules is enhanced, and lung fibrosis is promoted, but the mechanism for this is unclear. It has been reported that a downregulation of the Na,K­ATPase ß1 subunit in renal epithelial cells is involved in renal fibrosis development, but the role of this protein in lung fibrosis remains unknown. In the present study, the expression of the Na,K­ATPase ß1 subunit was revealed to be markedly decreased in AECs of patients with IPF and a bleomycin­induced pulmonary fibrosis mouse model. Treatment with transforming growth factor ß­1 led to significantly downregulation of the Na,K­ATPase ß1 subunit in lung adenocarcioma A549 cells. Furthermore, the knockdown of the Na,K­ATPase ß1 subunit in A549 cells resulted in the upregulation of profibrotic molecules, activation of the neurogenic locus notch homolog protein 1 and extracellular signal­regulated kinase 1/2 signaling pathways and induction of endoplasmic reticulum stress. These findings reveal that the downregulation of the Na,K­ATPase ß1 subunit enhances the expression of profibrotic molecules in AECs and may contribute to IPF pathogenesis.


Asunto(s)
Células Epiteliales Alveolares/enzimología , Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Fibrosis Pulmonar Idiopática/enzimología , Sistema de Señalización de MAP Quinasas , ATPasa Intercambiadora de Sodio-Potasio/biosíntesis , Células A549 , Adulto , Células Epiteliales Alveolares/patología , Animales , Estrés del Retículo Endoplásmico , Humanos , Fibrosis Pulmonar Idiopática/patología , Masculino , Ratones , Persona de Mediana Edad
19.
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
20.
J Cyst Fibros ; 18(3): e19-e25, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30415947

RESUMEN

BACKGROUND: Glycosaminoglycans (GAGs) are essential in many infections, including recurrent bacterial respiratory infections, the main cause of mortality in cystic fibrosis (CF) patients. METHODS: Using a cellular model of healthy and CF lung epithelium, a comparative transcriptomic study of GAG encoding genes was performed using qRT-PCR, and their differential involvement in the adhesion of bacterial pathogens analyzed by enzymatic degradation and binding competition experiments. RESULTS: Various alterations in gene expression in CF cells were found which affect GAG structures and seem to influence bacterial adherence to lung epithelium cells. Heparan sulfate appears to be the most important GAG species involved in bacterial binding. CONCLUSIONS: Adherence to lung epithelial cells of some of the main pathogens involved in CF is dependent on GAGs, and the expression of these polysaccharides is altered in CF cells, suggesting it could play an essential role in the development of infectious pathology.


Asunto(s)
Bacterias , Adhesión Bacteriana/fisiología , Sulfatos de Condroitina , Fibrosis Quística , Heparitina Sulfato , Infecciones del Sistema Respiratorio , Células Epiteliales Alveolares/enzimología , Bacterias/clasificación , Bacterias/metabolismo , Línea Celular , Sulfatos de Condroitina/biosíntesis , Sulfatos de Condroitina/metabolismo , Fibrosis Quística/metabolismo , Fibrosis Quística/microbiología , Perfilación de la Expresión Génica , Glicosaminoglicanos/fisiología , Heparitina Sulfato/biosíntesis , Heparitina Sulfato/metabolismo , Humanos , Infecciones del Sistema Respiratorio/metabolismo , Infecciones del Sistema Respiratorio/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...