Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros











Intervalo de año de publicación
1.
Thorax ; 77(2): 203-209, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34404754

RESUMEN

COVID-19 has different clinical stages, and effective therapy depends on the location and extent of the infection. The purpose of this review is to provide a background for understanding the progression of the disease throughout the pulmonary epithelium and discuss therapeutic options. The prime sites for infection that will be contrasted in this review are the conducting airways and the gas exchange portions of the lung. These two sites are characterised by distinct cellular composition and innate immune responses, which suggests the use of distinct therapeutic agents. In the nose, ciliated cells are the primary target cells for SARS-CoV-2 viral infection, replication and release. Infected cells shed their cilia, which disables mucociliary clearance. Evidence further points to a suppressed or incompletely activated innate immune response to SARS-CoV-2 infection in the upper airways. Asymptomatic individuals can still have a productive viral infection and infect others. In the gas exchange portion of the lung, the alveolar type II epithelial cell is the main target cell type. Cell death and marked innate immune response during infection likely contribute to alveolar damage and resultant acute respiratory distress syndrome. Alveolar infection can precipitate a hyperinflammatory state, which is the target of many therapies in severe COVID-19. Disease resolution in the lung is variable and may include scaring and long-term sequalae because the alveolar type II cells are also progenitor cells for the alveolar epithelium.


Asunto(s)
COVID-19 , Células Epiteliales , Humanos , Pulmón , Mucosa Respiratoria , SARS-CoV-2
2.
Am J Physiol Lung Cell Mol Physiol ; 319(1): L115-L120, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32493030

RESUMEN

COVID-19 can be divided into three clinical stages, and one can speculate that these stages correlate with where the infection resides. For the asymptomatic phase, the infection mostly resides in the nose, where it elicits a minimal innate immune response. For the mildly symptomatic phase, the infection is mostly in the pseudostratified epithelium of the larger airways and is accompanied by a more vigorous innate immune response. In the conducting airways, the epithelium can recover from the infection, because the keratin 5 basal cells are spared and they are the progenitor cells for the bronchial epithelium. There may be more severe disease in the bronchioles, where the club cells are likely infected. The devastating third phase is in the gas exchange units of the lung, where ACE2-expressing alveolar type II cells and perhaps type I cells are infected. The loss of type II cells results in respiratory insufficiency due to the loss of pulmonary surfactant, alveolar flooding, and possible loss of normal repair, since type II cells are the progenitors of type I cells. The loss of type I and type II cells will also block normal active resorption of alveolar fluid. Subsequent endothelial damage leads to transudation of plasma proteins, formation of hyaline membranes, and an inflammatory exudate, characteristic of ARDS. Repair might be normal, but if the type II cells are severely damaged alternative pathways for epithelial repair may be activated, which would result in some residual lung disease.


Asunto(s)
Células Epiteliales Alveolares/virología , Betacoronavirus/patogenicidad , Infecciones por Coronavirus/virología , Células Epiteliales/virología , Neumonía Viral/virología , Células Epiteliales Alveolares/metabolismo , COVID-19 , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/terapia , Células Epiteliales/metabolismo , Epitelio/metabolismo , Epitelio/virología , Humanos , Pulmón/metabolismo , Pandemias , Neumonía Viral/diagnóstico , Neumonía Viral/terapia , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/virología , SARS-CoV-2
4.
Cell Death Dis ; 10(9): 638, 2019 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-31474749

RESUMEN

DJ-1 is a multifunctional protein with cytoprotective functions. It is localized in the cytoplasm, nucleus, and mitochondria. The conserved cysteine residue at position 106 (Cys106) within DJ-1 serves as a sensor of redox state and can be oxidized to both the sulfinate (-SO2-) and sulfonate (-SO3-) forms. DJ-1 with Cys106-SO2- has cytoprotective activity but high levels of reactive oxygen species can induce its overoxidation to Cys106-SO3-. We found increased oxidative stress in alveolar type II (ATII) cells isolated from emphysema patients as determined by 4-HNE expression. DJ-1 with Cys106-SO3- was detected in these cells by mass spectrometry analysis. Moreover, ubiquitination of Cys106-SO3- DJ-1 was identified, which suggests that this oxidized isoform is targeted for proteasomal destruction. Furthermore, we performed controlled oxidation using H2O2 in A549 cells with DJ-1 knockout generated using CRISPR-Cas9 strategy. Lack of DJ-1 sensitized cells to apoptosis induced by H2O2 as detected using Annexin V and propidium iodide by flow cytometry analysis. This treatment also decreased both mitochondrial DNA amount and mitochondrial ND1 (NADH dehydrogenase 1, subunit 1) gene expression, as well as increased mitochondrial DNA damage. Consistent with the decreased cytoprotective function of overoxidized DJ-1, recombinant Cys106-SO3- DJ-1 exhibited a loss of its thermal unfolding transition, mild diminution of secondary structure in CD spectroscopy, and an increase in picosecond-nanosecond timescale dynamics as determined using NMR. Altogether, our data indicate that very high oxidative stress in ATII cells in emphysema patients induces DJ-1 overoxidation to the Cys106-SO3- form, leading to increased protein flexibility and loss of its cytoprotective function, which may contribute to this disease pathogenesis.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Cisteína/metabolismo , Proteína Desglicasa DJ-1/metabolismo , Anciano , Línea Celular Tumoral , Femenino , Humanos , Masculino , Persona de Mediana Edad , Oxidación-Reducción , Estrés Oxidativo/fisiología , Transfección
5.
Am J Physiol Lung Cell Mol Physiol ; 317(2): L283-L294, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31166130

RESUMEN

Epithelial-fibroblast interactions are thought to be very important in the adult lung in response to injury, but the specifics of these interactions are not well defined. We developed coculture systems to define the interactions of adult human alveolar epithelial cells with lung fibroblasts. Alveolar type II cells cultured on floating collagen gels reduced the expression of type 1 collagen (COL1A1) and α-smooth muscle actin (ACTA2) in fibroblasts. They also reduced fibroblast expression of hepatocyte growth factor (HGF), fibroblast growth factor 7 (FGF7, KGF), and FGF10. When type II cells were cultured at an air-liquid interface to maintain high levels of surfactant protein expression, this inhibitory activity was lost. When type II cells were cultured on collagen-coated tissue culture wells to reduce surfactant protein expression further and increase the expression of some type I cell markers, the epithelial cells suppressed transforming growth factor-ß (TGF-ß)-stimulated ACTA2 and connective tissue growth factor (CTGF) expression in lung fibroblasts. Our results suggest that transitional alveolar type II cells and likely type I cells but not fully differentiated type II cells inhibit matrix and growth factor expression in fibroblasts. These cells express markers of both type II cells and type I cells. This is probably a normal homeostatic mechanism to inhibit the fibrotic response in the resolution phase of wound healing. Defining how transitional type II cells convert activated fibroblasts into a quiescent state and inhibit the effects of TGF-ß may provide another approach to limiting the development of fibrosis after alveolar injury.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Células Epiteliales Alveolares/efectos de los fármacos , Células Cultivadas , Colágeno/farmacología , Células Epiteliales/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Surfactantes Pulmonares/metabolismo
6.
Sci Rep ; 9(1): 920, 2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30696938

RESUMEN

Emphysema is characterized by alveolar wall destruction induced mainly by cigarette smoke. Oxidative damage of DNA may contribute to the pathophysiology of this disease. We studied the impairment of the non-homologous end joining (NHEJ) repair pathway and DNA damage in alveolar type II (ATII) cells and emphysema development. We isolated primary ATII cells from control smokers, nonsmokers, and patients with emphysema to determine DNA damage and repair. We found higher reactive oxygen species generation and DNA damage in ATII cells obtained from individuals with this disease  in comparison with controls. We also observed low phosphorylation of H2AX, which activates DSBs repair signaling, in emphysema. Our results indicate the impairement  of NHEJ, as detected by low XLF expression. We also analyzed the role of DJ-1, which has a cytoprotective activity. We detected DJ-1 and  XLF interaction in ATII cells in emphysema, which suggests the impairment of their function. Moreover, we found that DJ-1 KO mice are more susceptible to DNA damage induced by cigarette smoke. Our results suggest that oxidative DNA damage and ineffective the DSBs repair via the impaired NHEJ may contribute to ATII cell death in emphysema.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Reparación del ADN por Unión de Extremidades , Enfisema Pulmonar/etiología , Enfisema Pulmonar/metabolismo , Animales , Biomarcadores , Daño del ADN , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Técnica del Anticuerpo Fluorescente , Expresión Génica , Humanos , Ratones , Estrés Oxidativo , Unión Proteica , Enfisema Pulmonar/patología , Especies Reactivas de Oxígeno/metabolismo , Fumar/efectos adversos
7.
Physiol Rep ; 6(16): e13794, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30155985

RESUMEN

TGF beta is a multifunctional cytokine that is important in the pathogenesis of pulmonary fibrosis. The ability of TGF beta to stimulate smooth muscle actin and extracellular matrix gene expression in fibroblasts is well established. In this report, we evaluated the effect of TGF beta on the expression of HGF, FGF7 (KGF), and FGF10, important growth and survival factors for the alveolar epithelium. These growth factors are important for maintaining type II cells and for restoration of the epithelium after lung injury. Under conditions of normal serum supplementation or serum withdrawal TGF beta inhibited fibroblast expression of HGF, FGF7, and FGF10. We confirmed these observations with genome wide RNA sequencing of the response of control and IPF fibroblasts to TGF beta. In general, gene expression in IPF fibroblasts was similar to control fibroblasts. Reduced expression of HGF, FGF7, and FGF10 is another means whereby TGF beta impairs epithelial healing and promotes fibrosis after lung injury.


Asunto(s)
Fibroblastos/efectos de los fármacos , Fibrosis Pulmonar Idiopática/patología , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Factor de Crecimiento Transformador beta/farmacología , Anciano , Anciano de 80 o más Años , Células Cultivadas , Medio de Cultivo Libre de Suero , Femenino , Factor 10 de Crecimiento de Fibroblastos/biosíntesis , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 7 de Crecimiento de Fibroblastos/biosíntesis , Factor 7 de Crecimiento de Fibroblastos/genética , Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Factor de Crecimiento de Hepatocito/biosíntesis , Factor de Crecimiento de Hepatocito/genética , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Pulmón/metabolismo , Masculino , Persona de Mediana Edad , ARN Mensajero/genética , Factor de Crecimiento Transformador beta/fisiología
8.
Respir Res ; 19(1): 126, 2018 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-29940963

RESUMEN

BACKGROUND: Airway epithelial cells and alveolar macrophages (AMs) are the first line of defense in the lung during infection. Toll-like receptor (TLR) agonists have been extensively used to define the regulation of inflammation in these cells. However, previous studies were performed in non-paired airway epithelial cells and AMs. The major goal of our study was to compare the pro- and anti-inflammatory responses of paired human primary airway epithelial cells and AMs to TLR3 and TLR4 agonists. METHODS: Tracheobronchial epithelial cells (TBEC) and AMs from four smokers and four non-smokers without lung disease were cultured with or without Poly(I:C) (PIC) (a TLR3 agonist) or LPS (a TLR4 agonist) for 4, 24 and 48 h. The immune responses of paired cells were compared. RESULTS: TBEC and AMs showed stronger pro-inflammatory cytokine (e.g., IL-8) responses to PIC and LPS, respectively. TLR3 and TLR4 mRNA levels were similar in non-stimulated TBEC and AMs. However, PIC stimulation in AMs led to sustained up-regulation of the immune negative regulators Tollip and A20, which may render AMs less sensitive to PIC stimulation than TBEC. Unlike AMs, TBEC did not increase NF-κB activation after LPS stimulation. Interestingly, smoking status was correlated with less TLR3 and IRAK-M expression in non-stimulated TBEC, but not in AMs. PIC-stimulated TBEC and LPS-stimulated AMs from smokers vs. non-smokers produced more IL-8. Finally, we show that expression of A20 and IRAK-M is strongly correlated in the two paired cell types. CONCLUSIONS: By using paired airway epithelial cells and AMs, this study reveals how these two critical types of lung cells respond to viral and bacterial pathogen associated molecular patterns, and provides rationale for modulating immune negative regulators to prevent excessive lung inflammation during respiratory infection.


Asunto(s)
Antiinflamatorios/inmunología , Mediadores de Inflamación/inmunología , Macrófagos Alveolares/inmunología , Mucosa Respiratoria/inmunología , Anciano , Antiinflamatorios/metabolismo , Células Cultivadas , Femenino , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/metabolismo , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Masculino , Persona de Mediana Edad , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Fumar/inmunología , Fumar/metabolismo
9.
Am J Pathol ; 187(8): 1772-1786, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28618253

RESUMEN

During the acute respiratory distress syndrome, epithelial cells, primarily alveolar type (AT) I cells, die and slough off, resulting in enhanced permeability. ATII cells proliferate and spread onto the denuded basement membrane to reseal the barrier. Repair of the alveolar epithelium is critical for clinical recovery; however, mechanisms underlying ATII cell proliferation and spreading are not well understood. We hypothesized that hypoxia-inducible factor (HIF)1α promotes proliferation and spreading of ATII cells during repair after lung injury. Mice were treated with lipopolysaccharide or hydrochloric acid. HIF activation in ATII cells after injury was demonstrated by increased luciferase activity in oxygen degradation domain-Luc (HIF reporter) mice and expression of the HIF1α target gene GLUT1. ATII cell proliferation during repair was attenuated in ATII cell-specific HIF1α knockout (SftpcCreERT2+/-;HIF1αf/f) mice. The HIF target vascular endothelial growth factor promoted ATII cell proliferation in vitro and after lung injury in vivo. In the scratch wound assay of cell spreading, HIF stabilization accelerated, whereas HIF1α shRNA delayed wound closure. SDF1 and its receptor, CXCR4, were found to be HIF1α-regulated genes in ATII cells and were up-regulated during lung injury. Stromal cell-derived factor 1/CXCR4 inhibition impaired cell spreading and delayed the resolution of permeability after lung injury. We conclude that HIF1α is activated in ATII cells after lung injury and promotes proliferation and spreading during repair.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Células Epiteliales Alveolares/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Alveolos Pulmonares/metabolismo , Transducción de Señal/fisiología , Animales , Línea Celular , Proliferación Celular/fisiología , Quimiocina CXCL12/metabolismo , Modelos Animales de Enfermedad , Ratones , Permeabilidad , Ratas , Receptores CXCR4/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas/fisiología
10.
Am J Respir Cell Mol Biol ; 55(3): 439-49, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27093578

RESUMEN

Cigarette smoke (CS) is a main source of oxidative stress and a key risk factor for emphysema, which consists of alveolar wall destruction. Alveolar type (AT) II cells are in the gas exchange regions of the lung. We isolated primary ATII cells from deidentified organ donors whose lungs were not suitable for transplantation. We analyzed the cell injury obtained from nonsmokers, moderate smokers, and heavy smokers. DJ-1 protects cells from oxidative stress and induces nuclear erythroid 2-related factor-2 (Nrf2) expression, which activates the antioxidant defense system. In ATII cells isolated from moderate smokers, we found DJ-1 expression by RT-PCR, and Nrf2 and heme oxygenase (HO)-1 translocation by Western blotting and immunocytofluorescence. In ATII cells isolated from heavy smokers, we detected Nrf2 and HO-1 cytoplasmic localization. Moreover, we found high oxidative stress, as detected by 4-hydroxynonenal (4-HNE) (immunoblotting), inflammation by IL-8 and IL-6 levels by ELISA, and apoptosis by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay in ATII cells obtained from heavy smokers. Furthermore, we detected early DJ-1 and late Nrf2 expression after ATII cell treatment with CS extract. We also overexpressed DJ-1 by adenovirus construct and found that this restored Nrf2 and HO-1 expression and induced nuclear translocation in heavy smokers. Moreover, DJ-1 overexpression also decreased ATII cell apoptosis caused by CS extract in vitro. Our results indicate that DJ-1 activates the Nrf2-mediated antioxidant defense system. Furthermore, DJ-1 overexpression can restore the impaired Nrf2 pathway, leading to ATII cell protection in heavy smokers. This suggests a potential therapeutic strategy for targeting DJ-1 in CS-related lung diseases.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Citoprotección , Factor 2 Relacionado con NF-E2/metabolismo , Proteína Desglicasa DJ-1/metabolismo , Fumar/efectos adversos , Adenoviridae/metabolismo , Aldehídos/metabolismo , Células Epiteliales Alveolares/patología , Apoptosis/genética , Separación Celular , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Proteína Desglicasa DJ-1/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
11.
Am J Physiol Lung Cell Mol Physiol ; 308(11): L1178-88, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26033355

RESUMEN

The most severe complication of influenza is viral pneumonia, which can lead to the acute respiratory distress syndrome. Alveolar epithelial cells (AECs) are the first cells that influenza virus encounters upon entering the alveolus. Infected epithelial cells produce cytokines that attract and activate neutrophils and macrophages, which in turn induce damage to the epithelial-endothelial barrier. Hepatocyte growth factor (HGF)/c-Met and transforming growth factor-α (TGF-α)/epidermal growth factor receptor (EGFR) are well known to regulate repair of damaged alveolar epithelium by stimulating cell migration and proliferation. Recently, TGF-α/EGFR signaling has also been shown to regulate innate immune responses in bronchial epithelial cells. However, little is known about whether HGF/c-Met signaling alters the innate immune responses and whether the innate immune responses in AECs are regulated by HGF/c-Met and TGF-α/EGFR. We hypothesized that HGF/c-Met and TGF-α/EGFR would regulate innate immune responses to influenza A virus infection in human AECs. We found that recombinant human HGF (rhHGF) and rhTGF-α stimulated primary human AECs to secrete IL-8 and granulocyte macrophage colony-stimulating factor (GM-CSF) strongly and IL-6 and monocyte chemotactic protein 1 moderately. Influenza infection stimulated the secretion of IL-8 and GM-CSF by AECs plated on rat-tail collagen through EGFR activation likely by TGF-α released from AECs and through c-Met activated by HGF secreted from lung fibroblasts. HGF secretion by fibroblasts was stimulated by AEC production of prostaglandin E2 during influenza infection. We conclude that HGF/c-Met and TGF-α/EGFR signaling enhances the innate immune responses by human AECs during influenza infections.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Interleucina-8/metabolismo , Células Epiteliales Alveolares/virología , Células Cultivadas , Técnicas de Cocultivo , Dinoprostona/metabolismo , Receptores ErbB/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Proteínas Proto-Oncogénicas c-met/metabolismo , Alveolos Pulmonares/patología , Transducción de Señal , Factor de Crecimiento Transformador alfa/metabolismo
12.
J Immunol ; 194(11): 5200-10, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25917096

RESUMEN

Collecting lymphatic vessels (CLVs), surrounded by fat and endowed with contractile muscle and valves, transport lymph from tissues after it is absorbed into lymphatic capillaries. CLVs are not known to participate in immune responses. In this study, we observed that the inherent permeability of CLVs allowed broad distribution of lymph components within surrounding fat for uptake by adjacent macrophages and dendritic cells (DCs) that actively interacted with CLVs. Endocytosis of lymph-derived Ags by these cells supported recall T cell responses in the fat and also generated Ag-bearing DCs for emigration into adjacent lymph nodes (LNs). Enhanced recruitment of DCs to inflammation-reactive LNs significantly relied on adipose tissue DCs to maintain sufficient numbers of Ag-bearing DCs as the LN expanded. Thus, CLVs coordinate inflammation and immunity within adipose depots and foster the generation of an unexpected pool of APCs for Ag transport into the adjacent LN.


Asunto(s)
Tejido Adiposo/inmunología , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Ganglios Linfáticos/inmunología , Vasos Linfáticos/metabolismo , Tejido Adiposo/patología , Animales , Movimiento Celular/inmunología , Células Dendríticas/metabolismo , Endocitosis , Humanos , Inflamación/inmunología , Ganglios Linfáticos/citología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Permeabilidad , Ratas , Ratas Sprague-Dawley , Linfocitos T/inmunología , Uniones Estrechas/inmunología
13.
Am J Physiol Lung Cell Mol Physiol ; 308(2): L199-207, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25326580

RESUMEN

The receptor tyrosine kinase human epidermal growth factor receptor-2 (HER2) is known to regulate pulmonary epithelial barrier function; however, the mechanisms behind this effect remain unidentified. We hypothesized that HER2 signaling alters the epithelial barrier through an interaction with the adherens junction (AJ) protein ß-catenin, leading to dissolution of the AJ. In quiescent pulmonary epithelial cells, HER2 and ß-catenin colocalized along the lateral intercellular junction. HER2 activation by the ligand neuregulin-1 was associated with tyrosine phosphorylation of ß-catenin, dissociation of ß-catenin from E-cadherin, and decreased E-cadherin-mediated cell adhesion. All effects were blocked with the HER2 inhibitor lapatinib. ß-Catenin knockdown using shRNA significantly attenuated neuregulin-1-induced decreases in pulmonary epithelial resistance in vitro. Our data indicate that HER2 interacts with ß-catenin, leading to dissolution of the AJ, decreased cell-cell adhesion, and disruption of the pulmonary epithelial barrier.


Asunto(s)
Uniones Adherentes/metabolismo , Adhesión Celular/fisiología , Receptor ErbB-2/metabolismo , Mucosa Respiratoria/metabolismo , beta Catenina/metabolismo , Línea Celular , Impedancia Eléctrica , Activación Enzimática , Humanos , Lapatinib , Pulmón/fisiología , Neurregulina-1/metabolismo , Permeabilidad , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño , Receptor ErbB-2/antagonistas & inhibidores , Transducción de Señal , Uniones Estrechas/metabolismo , beta Catenina/genética
14.
Biochem Biophys Res Commun ; 452(4): 1091-7, 2014 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-25251473

RESUMEN

Alveolar type II (ATII) cells remain differentiated and express surfactant proteins when cultured at an air-liquid (A/L) interface. When cultured under submerged conditions, ATII cells dedifferentiate and change their gene expression profile. We have previously shown that gene expression under submerged conditions is regulated by hypoxia inducible factor (HIF) signaling due to focal hypoxia resulting from ATII cell metabolism. Herein, we sought to further define gene expression changes in ATII cells cultured under submerged conditions. We performed a genome wide microarray on RNA extracted from rat ATII cells cultured under submerged conditions for 24-48h after switching from an A/L interface. We found significant alterations in gene expression, including upregulation of the HIF target genes stanniocalcin-1 (STC1), tyrosine hydroxylase (Th), enolase (Eno) 2, and matrix metalloproteinase (MMP) 13, and we verified upregulation of these genes by RT-PCR. Because STC1, a highly evolutionarily conserved glycoprotein with anti-inflammatory, anti-apoptotic, anti-oxidant, and wound healing properties, is widely expressed in the lung, we further explored the potential functions of STC1 in the alveolar epithelium. We found that STC1 was induced by hypoxia and HIF in rat ATII cells, and this induction occurred rapidly and reversibly. We also showed that recombinant human STC1 (rhSTC1) enhanced cell motility with extended lamellipodia formation in alveolar epithelial cell (AEC) monolayers but did not inhibit the oxidative damage induced by LPS. We also confirmed that STC1 was upregulated by hypoxia and HIF in human lung epithelial cells. In this study, we have found that several HIF target genes including STC1 are upregulated in AECs by a submerged condition, that STC1 is regulated by hypoxia and HIF, that this regulation is rapidly and reversibly, and that STC1 enhances wound healing moderately in AEC monolayers. However, STC1 did not inhibit oxidative damage in rat AECs stimulated by LPS in vitro. Therefore, alterations in gene expression by ATII cells under submerged conditions including STC1 were largely induced by hypoxia and HIF, which may be relevant to our understanding of the pathogenesis of various lung diseases in which the alveolar epithelium is exposed to relative hypoxia.


Asunto(s)
Hipoxia de la Célula/fisiología , Células Epiteliales/metabolismo , Glicoproteínas/metabolismo , Factor 1 Inducible por Hipoxia/metabolismo , Alveolos Pulmonares/metabolismo , Animales , Células Cultivadas , Células Epiteliales/citología , Regulación de la Expresión Génica/fisiología , Masculino , Alveolos Pulmonares/citología , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/fisiología
15.
Bioinformatics ; 30(17): 2393-8, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24812339

RESUMEN

MOTIVATION: Non-small-cell lung cancer (NSCLC) is the leading cause of cancer death in the United States. Targeted tyrosine kinase inhibitors (TKIs) directed against the epidermal growth factor receptor (EGFR) have been widely and successfully used in treating NSCLC patients with activating EGFR mutations. Unfortunately, the duration of response is short-lived, and all patients eventually relapse by acquiring resistance mechanisms. RESULT: We performed an integrative systems biology approach to determine essential kinases that drive EGFR-TKI resistance in cancer cell lines. We used a series of bioinformatics methods to analyze and integrate the functional genetics screen and RNA-seq data to identify a set of kinases that are critical in survival and proliferation in these TKI-resistant lines. By connecting the essential kinases to compounds using a novel kinase connectivity map (K-Map), we identified and validated bosutinib as an effective compound that could inhibit proliferation and induce apoptosis in TKI-resistant lines. A rational combination of bosutinib and gefitinib showed additive and synergistic effects in cancer cell lines resistant to EGFR TKI alone. CONCLUSIONS: We have demonstrated a bioinformatics-driven discovery roadmap for drug repurposing and development in overcoming resistance in EGFR-mutant NSCLC, which could be generalized to other cancer types in the era of personalized medicine. AVAILABILITY AND IMPLEMENTATION: K-Map can be accessible at: http://tanlab.ucdenver.edu/kMap. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma de Pulmón de Células no Pequeñas/genética , Receptores ErbB/genética , Neoplasias Pulmonares/genética , Inhibidores de Proteínas Quinasas/farmacología , Compuestos de Anilina/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Línea Celular Tumoral , Biología Computacional , Resistencia a Antineoplásicos/genética , Gefitinib , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Mutación , Nitrilos/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Quinasas/genética , Quinazolinas/farmacología , Quinolinas/farmacología , Análisis de Secuencia de ARN
16.
Am J Physiol Lung Cell Mol Physiol ; 307(1): L94-105, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24748602

RESUMEN

There are 190,600 cases of acute lung injury/acute respiratory distress syndrome (ALI/ARDS) each year in the United States, and the incidence and mortality of ALI/ARDS increase dramatically with age. Patients with ALI/ARDS have alveolar epithelial injury, which may be worsened by high-pressure mechanical ventilation. Alveolar type II (ATII) cells are the progenitor cells for the alveolar epithelium and are required to reestablish the alveolar epithelium during the recovery process from ALI/ARDS. Lung fibroblasts (FBs) migrate and proliferate early after lung injury and likely are an important source of growth factors for epithelial repair. However, how lung FBs affect epithelial wound healing in the human adult lung has not been investigated in detail. Hepatocyte growth factor (HGF) is known to be released mainly from FBs and to stimulate both migration and proliferation of primary rat ATII cells. HGF is also increased in lung tissue, bronchoalveolar lavage fluid, and serum in patients with ALI/ARDS. Therefore, we hypothesized that HGF secreted by FBs would enhance wound closure in alveolar epithelial cells (AECs). Wound closure was measured using a scratch wound-healing assay in primary human AEC monolayers and in a coculture system with FBs. We found that wound closure was accelerated by FBs mainly through HGF/c-Met signaling. HGF also restored impaired wound healing in AECs from the elderly subjects and after exposure to cyclic stretch. We conclude that HGF is the critical factor released from FBs to close wounds in human AEC monolayers and suggest that HGF is a potential strategy for hastening alveolar repair in patients with ALI/ARDS.


Asunto(s)
Fibroblastos/citología , Factor de Crecimiento de Hepatocito/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Alveolos Pulmonares/citología , Cicatrización de Heridas/fisiología , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Movimiento Celular , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/patología , Fibroblastos/metabolismo , Humanos , Interleucina-1alfa/farmacología , Interleucina-1beta/farmacología , Seudópodos/fisiología , Transducción de Señal
17.
PLoS One ; 8(7): e70129, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23894604

RESUMEN

Because they are the natural target for respiratory pathogens, primary human respiratory epithelial cells provide the ideal in vitro system for isolation and study of human respiratory viruses, which display a high degree of cell, tissue, and host specificity. Human coronavirus HKU1, first discovered in 2005, has a worldwide prevalence and is associated with both upper and lower respiratory tract disease in both children and adults. Research on HCoV-HKU1 has been difficult because of its inability to be cultured on continuous cell lines and only recently it was isolated from clinical specimens using primary human, ciliated airway epithelial cells. Here we demonstrate that HCoV-HKU1 can infect and be serially propagated in primary human alveolar type II cells at the air-liquid interface. We were not able to infect alveolar type I-like cells or alveolar macrophages. Type II alveolar cells infected with HCoV-HKU1 demonstrated formation of large syncytium. At 72 hours post inoculation, HCoV-HKU1 infection of type II cells induced increased levels of mRNAs encoding IL29,CXCL10, CCL5, and IL-6 with no significant increases in the levels of IFNß. These studies demonstrate that type II cells are a target cell for HCoV-HKU1 infection in the lower respiratory tract, that type II alveolar cells are immune-competent in response to infection exhibiting a type III interferon and proinflammatory chemokine response, and that cell to cell spread may be a major factor for spread of infection. Furthermore, these studies demonstrate that human alveolar cells can be used to isolate and study novel human respiratory viruses that cause lower respiratory tract disease.


Asunto(s)
Infecciones por Coronavirus/inmunología , Coronavirus/inmunología , Inmunidad Innata/fisiología , Adolescente , Adulto , Células Cultivadas , Preescolar , Coronavirus/patogenicidad , Infecciones por Coronavirus/virología , Células Epiteliales/inmunología , Células Epiteliales/virología , Femenino , Humanos , Lactante , Interleucina-6/metabolismo , Macrófagos Alveolares/citología , Macrófagos Alveolares/metabolismo , Masculino , ARN Viral , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/metabolismo , Adulto Joven
18.
PLoS One ; 8(6): e67616, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23799150

RESUMEN

The 2009/2010 pandemic influenza virus (H1N1pdm) contains an avian-lineage PB2 gene that lacks E627K and D701N substitutions important in the pathogenesis and transmission of avian-origin viruses in humans or other mammals. Previous studies have shown that PB2-627K is not necessary because of a compensatory Q591R substitution. The role that PB2-701N plays in the H1N1pdm phenotype is not well understood. Therefore, PB2-D701N was introduced into an H1N1pdm virus (A/New York/1682/2009 (NY1682)) and analyzed in vitro and in vivo. Mini-genome replication assay, in vitro replication characteristics in cell lines, and analysis in the mouse and ferret models demonstrated that PB2-D701N increased virus replication rates and resulted in more severe pathogenicity in mice and more efficient transmission in ferrets. In addition, compared to the NY1682-WT virus, the NY1682-D701N mutant virus induced less IFN-λ and replicated to a higher titer in primary human alveolar epithelial cells. These findings suggest that the acquisition of the PB2-701N substitution by H1N1pdm viruses may result in more severe disease or increase transmission in humans.


Asunto(s)
Sustitución de Aminoácidos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/transmisión , ARN Polimerasa Dependiente del ARN/genética , Proteínas Virales/genética , Replicación Viral/genética , Células Epiteliales Alveolares/inmunología , Células Epiteliales Alveolares/virología , Animales , Asparagina/genética , Línea Celular Tumoral , Citocinas/metabolismo , Perros , Femenino , Hurones , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Gripe Humana/enzimología , Gripe Humana/virología , Cinética , Masculino , Ratones , Ratones Endogámicos BALB C
19.
Am J Respir Cell Mol Biol ; 48(5): 559-67, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23492188

RESUMEN

Emphysema is caused by the cigarette smoke (CS)-induced destruction of alveolar wall septa, and CS is the main risk factor for chronic obstructive pulmonary disease (COPD). To study the mechanisms of response to this insult, we focused on oxidant-induced lung injury and the potential role of nuclear erythroid 2-related factor-2 (Nrf2), which is a key regulator of the antioxidant defense system. We studied the protective role of N-acetylcysteine (NAC) against the injury of alveolar type II (ATII) cells induced by CS in vivo and in vitro. ATII cells were isolated and purified using magnetic MicroBeads (Miltenyi Biotec, Auburn, CA) from Nrf2(-/-) mice and wild-type mice. We analyzed pulmonary injury, inflammation, glutathione (GSH) concentrations, the expression of glutathione cysteine ligase catalytic subunit mRNA, glutathione cysteine ligase modifier subunit mRNA, and glutathione reductase mRNA, and Nrf2, heme oxygenase-1, and nicotinamide adenine dinucleotide phosphate-reduced:quinone oxireductase levels by Western blotting, TUNEL assay, and immunocytofluorescence for 4-hydroxynonenal as a marker of oxidative stress. We found that CS induced greater injury in ATII cells obtained from Nrf2(-/-) mice than from wild-type mice. Furthermore, NAC attenuated the injuries by CS in ATII cells obtained from wild-type mice both in vivo and in vitro. Moreover, NAC decreased the injury of ATII cells obtained from Nrf2(-/-) mice. Our results suggest that Nrf2-GSH signaling is important for the protective activity of NAC. In addition, in ATII cells deficient in Nrf2, this compound can provide partial protection through its reactive oxygen species-scavenging activities. Targeting the antioxidant system regulated by Nrf2 may provide an effective strategy against lung injury in COPD.


Asunto(s)
Acetilcisteína/farmacología , Células Epiteliales Alveolares/efectos de los fármacos , Depuradores de Radicales Libres/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Fumar/efectos adversos , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/fisiología , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citoprotección , Enfisema/tratamiento farmacológico , Enfisema/etiología , Enfisema/patología , Expresión Génica , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo , Humo , Nicotiana/química
20.
Am J Respir Cell Mol Biol ; 48(6): 742-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23418343

RESUMEN

Severe acute respiratory syndrome (SARS)-coronavirus (CoV) produces a devastating primary viral pneumonia with diffuse alveolar damage and a marked increase in circulating cytokines. One of the major cell types to be infected is the alveolar type II cell. However, the innate immune response of primary human alveolar epithelial cells infected with SARS-CoV has not been defined. Our objectives included developing a culture system permissive for SARS-CoV infection in primary human type II cells and defining their innate immune response. Culturing primary human alveolar type II cells at an air-liquid interface (A/L) improved their differentiation and greatly increased their susceptibility to infection, allowing us to define their primary interferon and chemokine responses. Viral antigens were detected in the cytoplasm of infected type II cells, electron micrographs demonstrated secretory vesicles filled with virions, virus RNA concentrations increased with time, and infectious virions were released by exocytosis from the apical surface of polarized type II cells. A marked increase was evident in the mRNA concentrations of interferon-ß and interferon-λ (IL-29) and in a large number of proinflammatory cytokines and chemokines. A surprising finding involved the variability of expression of angiotensin-converting enzyme-2, the SARS-CoV receptor, in type II cells from different donors. In conclusion, the cultivation of alveolar type II cells at an air-liquid interface provides primary cultures in which to study the pulmonary innate immune responses to infection with SARS-CoV, and to explore possible therapeutic approaches to modulating these innate immune responses.


Asunto(s)
Células Epiteliales/inmunología , Inmunidad Innata , Síndrome Respiratorio Agudo Grave/inmunología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , Enzima Convertidora de Angiotensina 2 , Antígenos Virales/inmunología , Antígenos Virales/metabolismo , Diferenciación Celular , Citoplasma/inmunología , Citoplasma/ultraestructura , Citoplasma/virología , Células Epiteliales/virología , Humanos , Interferón beta/inmunología , Interferón beta/metabolismo , Interferones , Interleucinas/inmunología , Interleucinas/metabolismo , Peptidil-Dipeptidasa A/inmunología , Peptidil-Dipeptidasa A/metabolismo , Cultivo Primario de Células , Alveolos Pulmonares/citología , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/virología , ARN Mensajero/metabolismo , Receptores Virales/metabolismo , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/virología , Factores de Tiempo , Liberación del Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA