Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Chinese Journal of Lung Cancer ; (12): 732-740, 2023.
Artículo en Chino | WPRIM | ID: wpr-1010081

RESUMEN

BACKGROUND@#Currently, a significant number of miners are involved in mining operations at the Gejiu tin mine in Yunnan. This occupational setting is associated with exposure to dust particles, heavy metals, polycyclic aromatic hydrocarbons, and radioactive radon, thereby significantly elevating the risk of lung cancer. This study aims to investigate the involvement of leptin-mediated extracellular regulated protein kinase (ERK) signaling pathway in the malignant transformation of rat alveolar type II epithelial cells induced by Yunnan tin mine dust.@*METHODS@#Immortalized rat alveolar cells type II (RLE-6TN) cells were infected with Yunnan tin mine dust at a concentration of 200 μg/mL for nine consecutive generations to establish the infected cell model, which was named R₂₀₀ cells. The cells were cultured normally, named as R cells. The expression of leptin receptor in both cell groups was detected using the Western blot method. The optimal concentration of leptin and mitogen-activated protein kinase kinase (MEK) inhibitor (U0126) on R₂₀₀ cells was determined using the MTT method. Starting from the 20th generation, the cells in the R group were co-cultured with leptin, while the cells in the R₂₀₀ group were co-cultured with the MEK inhibitor U0126. The morphological alterations of the cells in each group were visualized utilizing hematoxylin-eosin staining. Additionally, concanavalin A (ConA) was utilized to detect any morphological differences, and an anchorage-independent growth assay was conducted to assess the malignant transformation of the cells. The changes in the ERK signaling pathway in epithelial cells after the action of leptin were detected using the Western blot method.@*RESULTS@#Both the cells in the R group and R₂₀₀ group express leptin receptor OB-R. Compared to the R₂₀₀ group, the concentration of leptin at 100 ng/mL shows the most significant pro-proliferation effect. The proliferation of R₂₀₀ cells infected with the virus is inhibited by 30 μmol/L U0126, and a statistically significant divergence was seen when compared to the control group (P<0.05). Starting from the 25th generation, the cell morphology of the leptin-induced R₂₀₀ group (R₂₀₀L group) underwent changes, leading to malignant transformation observed at the 30th generation. The characteristics of malignant transformation became evident by the 40th generation in the R₂₀₀L group. In contrast, the other groups showed agglutination of P40 cells, and the speed of cell aggregation increased with an increase in ConA concentration. Notably, the R₂₀₀L group exhibited faster cell aggregation compared to the U0126-induced R₂₀₀ (R₂₀₀LU) group. Additionally, the cells in the R₂₀₀L group were capable of forming clones starting from P30, with a colony formation rate of 2.25‰±0.5‰. However, no clonal colonies were observed in the R₂₀₀LU group and R₂₀₀ group. The expression of phosphorylated extracellular signal-regulated kinase (pERK) was enhanced in cells of the R₂₀₀L group. However, when the cells in the R₂₀₀L group were treated with U0126, a blocking agent, the phosphorylation level of pERK decreased.@*CONCLUSIONS@#Leptin can promote the malignant transformation of lung epithelial cells infected by mine dust, and the ERK signaling pathway may be necessary for the transformation of alveolar type II epithelial cells induced by Yunnan tin mine dust.


Asunto(s)
Ratas , Animales , Células Epiteliales Alveolares/patología , Polvo , Estaño/efectos adversos , Neoplasias Pulmonares/patología , Leptina/efectos adversos , Receptores de Leptina , China , Transducción de Señal , Células Epiteliales/patología , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos adversos
2.
Chinese Journal of Traumatology ; (6): 155-161, 2023.
Artículo en Inglés | WPRIM | ID: wpr-981928

RESUMEN

PURPOSE@#This study aims to elucidate the electrotaxis response of alveolar epithelial cells (AECs) in direct-current electric fields (EFs), explore the impact of EFs on the cell fate of AECs, and lay the foundation for future exploitation of EFs for the treatment of acute lung injury.@*METHODS@#AECs were extracted from rat lung tissues using magnetic-activated cell sorting. To elucidate the electrotaxis responses of AECs, different voltages of EFs (0, 50, 100, and 200 mV/mm) were applied to two types of AECs, respectively. Cell migrations were recorded and trajectories were pooled to better demonstrate cellular activities through graphs. Cell directionality was calculated as the cosine value of the angle formed by the EF vector and cell migration. To further demonstrate the impact of EFs on the pulmonary tissue, the human bronchial epithelial cells transformed with Ad12-SV40 2B (BEAS-2B cells) were obtained and experimented under the same conditions as AECs. To determine the influence on cell fate, cells underwent electric stimulation were collected to perform Western blot analysis.@*RESULTS@#The successful separation and culturing of AECs were confirmed through immunofluorescence staining. Compared with the control, AECs in EFs demonstrated a significant directionality in a voltage-dependent way. In general, type Ⅰ alveolar epithelial cells migrated faster than type Ⅱ alveolar epithelial cells, and under EFs, these two types of cells exhibited different response threshold. For type Ⅱ alveolar epithelial cells, only EFs at 200 mV/mm resulted a significant difference to the velocity, whereas for, EFs at both 100 mV/mm and 200 mV/mm gave rise to a significant difference. Western blotting suggested that EFs led to an increased expression of a AKT and myeloid leukemia 1 and a decreased expression of Bcl-2-associated X protein and Bcl-2-like protein 11.@*CONCLUSION@#EFs could guide and accelerate the directional migration of AECs and exert antiapoptotic effects, which indicated that EFs are important biophysical signals in the re-epithelialization of alveolar epithelium in lung injury.


Asunto(s)
Humanos , Ratas , Animales , Células Epiteliales Alveolares , Pulmón , Lesión Pulmonar , Movimiento Celular/fisiología
3.
Chinese Journal of Natural Medicines (English Ed.) ; (6): 576-588, 2023.
Artículo en Inglés | WPRIM | ID: wpr-1010971

RESUMEN

Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is characterized by diffuse alveolar injury primarily caused by an excessive inflammatory response. Regrettably, the lack of effective pharmacotherapy currently available contributes to the high mortality rate in patients with this condition. Xuebijing (XBJ), a traditional Chinese medicine recognized for its potent anti-inflammatory properties, exhibits promise as a potential therapeutic agent for ALI/ARDS. This study aimed to explore the preventive effects of XBJ on ALI and its underlying mechanism. To this end, we established an LPS-induced ALI model and treated ALI mice with XBJ. Our results demonstrated that pre-treatment with XBJ significantly alleviated lung inflammation and increased the survival rate of ALI mice by 37.5%. Moreover, XBJ substantially suppressed the production of TNF-α, IL-6, and IL-1β in the lung tissue. Subsequently, we performed a network pharmacology analysis and identified identified 109 potential target genes of XBJ that were mainly involved in multiple signaling pathways related to programmed cell death and anti-inflammatory responses. Furthermore, we found that XBJ exerted its inhibitory effect on gasdermin-E-mediated pyroptosis of lung cells by suppressing TNF-α production. Therefore, this study not only establishes the preventive efficacy of XBJ in ALI but also reveals its role in protecting alveolar epithelial cells against gasdermin-E-mediated pyroptosis by reducing TNF-α release.


Asunto(s)
Animales , Ratones , Células Epiteliales Alveolares , Piroptosis , Gasderminas , Lipopolisacáridos/efectos adversos , Factor de Necrosis Tumoral alfa , Lesión Pulmonar Aguda/tratamiento farmacológico , Síndrome de Dificultad Respiratoria del Recién Nacido
4.
Braz. J. Pharm. Sci. (Online) ; 59: e22476, 2023. graf
Artículo en Inglés | LILACS | ID: biblio-1505847

RESUMEN

Abstract The aim of the present study was to investigate the effect of swertiamarin (STM) in attenuating paraquat (PQ)-induced human lung alveolar epithelial-like cell (A549) apoptosis and the underlying mechanisms. A549 cells were pretreated with different concentrations of STM for 2 hr and then cultured with or without PQ (700 µM) for 24 hr. Cell survival was determined using the CCK8 assay. Morphological changes, MDA content, inflammatory factors, fibrogenesis parameters, apoptosis rates, redox status and mitochondrial membrane potential (MMP) were evaluated. The expression of several genes involved in the modulation of redox status was measured by Western blotting. Cell viability and MMP were decreased, but the apoptosis rate and DCFH oxidation were elevated by PQ exposure. STM pretreatment notably increased cell viability and MMP and reduced the apoptosis rate and DCFH oxidation. Furthermore, TLR4- NOX4 signaling was significantly inhibited by STM. The downregulation of NOX4 by siRNA exerted the same protective effects as STM. This study provides the first evidence that STM attenuates PQ-induced pulmonary epithelial-like cell apoptosis via NOX4-mediated regulation of redox and mitochondrial function


Asunto(s)
Paraquat/efectos adversos , Células Epiteliales Alveolares/clasificación , ARN Interferente Pequeño/agonistas , NADPH Oxidasa 4/efectos adversos
5.
Chinese Journal of Traumatology ; (6): 138-144, 2022.
Artículo en Inglés | WPRIM | ID: wpr-928493

RESUMEN

PURPOSE@#The incidence of acute lung injury (ALI) in severe trauma patients is 48% and the mortality rate following acute respiratory distress syndrome evolved from ALI is up to 68.5%. Alveolar epithelial type 1 cells (AEC1s) and type 2 cells (AEC2s) are the key cells in the repair of injured lungs as well as fetal lung development. Therefore, the purification and culture of AEC1s and AEC2s play an important role in the research of repair and regeneration of lung tissue.@*METHODS@#Sprague-Dawley rats (3-4 weeks, 120-150 g) were purchased for experiment. Dispase and DNase I were jointly used to digest lung tissue to obtain a single-cell suspension of whole lung cells, and then magnetic bead cell sorting was performed to isolate T1α positive cells as AEC1s from the single-cell suspension by using polyclonal rabbit anti-T1a (a specific AEC1s membrane protein) antibodies combined with anti-rabbit IgG microbeads. Afterwards, alveolar epithelial cell membrane marker protein EpCAM was designed as a key label to sort AEC2s from the remaining T1α-neg cells by another positive immunomagnetic selection using monoclonal mouse anti-EpCAM antibodies and anti-mouse IgG microbeads. Cell purity was identified by immunofluorescence staining and flow cytometry.@*RESULTS@#The purity of AEC1s and AEC2s was 88.3% ± 3.8% and 92.6% ± 2.7%, respectively. The cell growth was observed as follows: AEC1s stretched within the 12-16 h, but the cells proliferated slowly; while AEC2s began to stretch after 24 h and proliferated rapidly from the 2nd day and began to differentiate after 3 days.@*CONCLUSION@#AEC1s and AEC2s sorted by this method have high purity and good viability. Therefore, our method provides a new approach for the isolation and culture of AEC1s and AEC2s as well as a new strategy for the research of lung repair and regeneration.


Asunto(s)
Animales , Ratas , Células Epiteliales Alveolares/citología , Técnicas de Cultivo de Célula , Separación Celular/métodos , Inmunoglobulina G/metabolismo , Pulmón , Fenómenos Magnéticos , Ratas Sprague-Dawley
6.
Protein & Cell ; (12): 717-733, 2021.
Artículo en Inglés | WPRIM | ID: wpr-888715

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic is caused by infection with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is spread primary via respiratory droplets and infects the lungs. Currently widely used cell lines and animals are unable to accurately mimic human physiological conditions because of the abnormal status of cell lines (transformed or cancer cells) and species differences between animals and humans. Organoids are stem cell-derived self-organized three-dimensional culture in vitro and model the physiological conditions of natural organs. Here we showed that SARS-CoV-2 infected and extensively replicated in human embryonic stem cells (hESCs)-derived lung organoids, including airway and alveolar organoids which covered the complete infection and spread route for SARS-CoV-2 within lungs. The infected cells were ciliated, club, and alveolar type 2 (AT2) cells, which were sequentially located from the proximal to the distal airway and terminal alveoli, respectively. Additionally, RNA-seq revealed early cell response to virus infection including an unexpected downregulation of the metabolic processes, especially lipid metabolism, in addition to the well-known upregulation of immune response. Further, Remdesivir and a human neutralizing antibody potently inhibited SARS-CoV-2 replication in lung organoids. Therefore, human lung organoids can serve as a pathophysiological model to investigate the underlying mechanism of SARS-CoV-2 infection and to discover and test therapeutic drugs for COVID-19.


Asunto(s)
Humanos , Adenosina Monofosfato/uso terapéutico , Alanina/uso terapéutico , Células Epiteliales Alveolares/virología , Anticuerpos Neutralizantes/uso terapéutico , COVID-19/virología , Regulación hacia Abajo , Descubrimiento de Drogas , Células Madre Embrionarias Humanas/metabolismo , Inmunidad , Metabolismo de los Lípidos , Pulmón/virología , ARN Viral/metabolismo , SARS-CoV-2/fisiología , Replicación Viral/efectos de los fármacos
7.
China Journal of Chinese Materia Medica ; (24): 146-154, 2021.
Artículo en Chino | WPRIM | ID: wpr-878923

RESUMEN

This study aimed to assess whether chrysin(ChR) can inhibit epithelial-mesenchymal transition(EMT) of type Ⅱ alveolar epithelial cell and produce anti-pulmonary fibrosis effect by regulating the NF-κB/Twist 1 signaling pathway. Sixty rats were randomly divided into the control group, the bleomycin(BLC) group, BLC+ChR(50 mg·kg~(-1)) group and BLC+ChR(100 mg·kg~(-1)) group, with 15 rats in each group. The pulmonary fibrosis model was induced by intratracheal injection of BLC(7 500 U·kg~(-1)). Rats were orally administered with different doses of ChR after BLC injection for 28 days. The cells were divided into control group, TGF-β1 group(5 ng·mL~(-1)), and TGF-β1+ChR(1, 10, 100 μmol·L~(-1)) groups. The type Ⅱ alveolar epithelial cells were treated with TGF-β1 for 24 h, and then treated with TGF-β1 for 48 h in the presence or absence of different doses of ChR(1, 10 and 100 μmol·L~(-1)). The morphological changes and collagen deposition in lung tissues were analyzed by HE staining, Masson staining and immunohistochemistry. The mRNA and protein expression levels of collagen Ⅰ, E-cadherin, zonula occludens-1(ZO-1), vimentin, alpha smooth muscle actin(α-SMA), inhibitor of nuclear factor kappa B alpha(IκBα), nuclear factor-kappa B p65(NF-κB p65), phospho-NF-κB p65(p-p65) and Twist 1 in lung tissues and cells were detected by qPCR and Western blot, respectively. The animal experiment results showed that as compared with the BLC group, after administration of ChR for 28 days, bleomycin-induced pulmonary fibrosis in rats was significantly relieved, collagen Ⅰ expression in lung tissues was significantly reduced(P<0.05 or P<0.01), and EMT of alveolar epithelial cells was obviously inhibited [the expression levels of E-cadherin and ZO-1 were increased and the expression levels of vimentin and α-SMA were decreased(P<0.05 or P<0.01)], concomitantly with significantly reduced IκBα and p65 phosphorylation level in cytoplasm and decreased NF-κB p65 and Twist 1 expression in nucleus(P<0.05 or P<0.01). The cell experiment results showed that different doses of ChR(1, 10 and 100 μmol·L~(-1)) significantly reduced TGF-β1-induced collagen Ⅰ expression(P<0.05 or P<0.01), significantly inhibited EMT of type Ⅱ alveolar epithelial cells[the expression levels of E-cadherin and ZO-1 were increased and the expression levels of vimentin and α-SMA were decreased(P<0.05 or P<0.01)], and inhibited IκBα and p65 phosphorylation in cytoplasm and down-regulated NF-κB p65 and Twist 1 expression in nucleus induced by TGF-β1(P<0.05 or P<0.01). The results suggest that ChR can reverse EMT of type Ⅱ alveolar epithelial cell and alleviate pulmonary fibrosis in rats, and its mechanism may be associated with reducing IκBα phosphorylation and inhibiting NF-κB p65 phosphorylation and nuclear transfer, thus down-regulating Twist 1 expression.


Asunto(s)
Animales , Ratas , Células Epiteliales Alveolares/metabolismo , Transición Epitelial-Mesenquimal , Flavonoides , FN-kappa B/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/genética
8.
Chinese Medical Journal ; (24): 261-274, 2021.
Artículo en Inglés | WPRIM | ID: wpr-878043

RESUMEN

There have been recent extensive studies and rapid advancement on the pathogenesis underlying idiopathic pulmonary fibrosis (IPF), and intricate pathogenesis of IPF has been suggested. The purpose of this study was to clarify the logical relationship between these mechanisms. An extensive search was undertaken of the PubMed using the following keywords: "etiology," "pathogenesis," "alveolar epithelial cell (AEC)," "fibroblast," "lymphocyte," "macrophage," "epigenomics," "histone," acetylation," "methylation," "endoplasmic reticulum stress," "mitochondrial dysfunction," "telomerase," "proteases," "plasminogen," "epithelial-mesenchymal transition," "oxidative stress," "inflammation," "apoptosis," and "idiopathic pulmonary fibrosis." This search covered relevant research articles published up to April 30, 2020. Original articles, reviews, and other articles were searched and reviewed for content; 240 highly relevant studies were obtained after screening. IPF is likely the result of complex interactions between environmental, genetic, and epigenetic factors: environmental exposures affect epigenetic marks; epigenetic processes translate environmental exposures into the regulation of chromatin; epigenetic processes shape gene expression profiles; in turn, an individual's genetic background determines epigenetic marks; finally, these genetic and epigenetic factors act in concert to dysregulate gene expression in IPF lung tissue. The pathogenesis of IPF involves various imbalances including endoplasmic reticulum, telomere length homeostasis, mitochondrial dysfunction, oxidant/antioxidant imbalance, Th1/Th2 imbalance, M1-M2 polarization of macrophages, protease/antiprotease imbalance, and plasminogen activation/inhibition imbalance. These affect each other, promote each other, and ultimately promote AEC/fibroblast apoptosis imbalance directly or indirectly. Excessive AEC apoptosis and impaired apoptosis of fibroblasts contribute to fibrosis. IPF is likely the result of complex interactions between environmental, genetic, and epigenetic factors. The pathogenesis of IPF involves various imbalances centered on AEC/fibroblast apoptosis imbalance.


Asunto(s)
Humanos , Células Epiteliales Alveolares , Apoptosis , Estrés del Retículo Endoplásmico , Fibroblastos , Fibrosis Pulmonar Idiopática/genética
9.
Rev. Ciênc. Méd. Biol. (Impr.) ; 19(4): 602-605, dez 30, 2020.
Artículo en Inglés | LILACS | ID: biblio-1355219

RESUMEN

Introduction: the severe acute respiratory syndrome ­ coronavirus 2 (SARS Cov-2), leads to a diffuse alveolar deterioration due infection of type II pneumocytes. The type II pneumocytes are involved in synthesis and secretion of pulmonary surfactant in pulmonary alveoli. Objective: the purpose of this study is to discuss the indication of surfactant replacement as a potential adjunctive treatment modality for SARS CoV-2, similarly treatment to neonatal respiratory distress syndrome. Methodology: we argue that SARS can be triggered by surfactant deficiency secondary to production deficiency determined by type 2 pneumocyte injuries. In this sense, we carried out a bibliographic review. Conclusion: thus, the replacement of human surfactant could be a potential treatment modality for SARS CoV-2, in the same way that it is indicated for the treatment of neonatal respiratory distress syndrome.


Introdução: a síndrome respiratória aguda grave ­ coronavírus 2 (SARS Cov-2), leva a uma deterioração alveolar difusa devido à infecção do pneumócitos tipo II. Os pneumócitos tipo II estão envolvidos na síntese e secreção de surfactante pulmonar nos alvéolos pulmonares. Objetivo: o objetivo deste estudo é discutir a indicação de reposição de surfactante como uma potencial modalidade de tratamento adjuvante para SARS CoV-2, similarmente ao tratamento da síndrome do desconforto respiratório neonatal. Metodologia: argumentamos que a SARS pode ser desencadeada pela deficiência de surfactante, secundária à deficiência da sua produção determinada por lesões de pneumócitos tipo 2. Nesse sentido, realizamos uma revisão bibliográfica. Conclusão: o uso de surfactante humana pode ser uma potencial modalidade de tratamento para a SARS CoV-2, da mesma forma que é indicada para o tratamento da síndrome do desconforto respiratório neonatal.


Asunto(s)
Surfactantes Pulmonares , Síndrome Respiratorio Agudo Grave , Células Epiteliales Alveolares , SARS-CoV-2 , Revisión , Informe Anual
10.
NOVA publ. cient ; 18(spe35): 11-33, jul.-dic. 2020. tab, graf
Artículo en Español | LILACS | ID: biblio-1149463

RESUMEN

Resumen El 31 de diciembre de 2019 la comisión municipal de salud de Wuhan (provincia de Hubei, China) informa sobre un inusitado brote de casos de neumonía en la ciudad. Posteriormente se determina que se trata de un nuevo coronavirus designado inicialmente como 2019-nCoV y posteriormente, SARS-CoV-2. El SARS-CoV-2 infecta y se replica en los neumocitos y macrófagos del sistema respiratorio específicamente en el parénquima pulmonar en donde reside el receptor celular ACE-2. Esta revisión describe aspectos relacionados con la transmisión, prevención, generalidades bioquímicas del SARS-CoV-2 y métodos diagnósticos del COVID-19. Inicialmente se describe la forma de transmisión del virus y algunas recomendaciones generales para su prevención. Posteriormente, se hace una descripción detallada de los aspectos bioquímicos del SARS-CoV-2, su ciclo infeccioso y la estructura de la proteína S, la cual está involucrada con el proceso de ingreso del virus a la célula. Finalmente, se describen los métodos y pruebas de laboratorio para el diagnóstico del COVID-19.


Abstract On December 31, 2019, Wuhan Municipal Health Commission (Hubei Province, China) reports on an unusual outbreak of pneumonia cases in the city. Subsequently it is determined that it is a new coronavirus initially designated as 2019-nCoV and later, SARS-CoV-2. SARS-CoV-2 infects and replicates in pneumocytes and macrophages of the respiratory system specifically in the lung parenchyma where the ACE-2 cell receptor resides. This review describes aspects related to the transmission, prevention, biochemical generalities of SARS-CoV-2 and diagnostic methods of COVID-19. Initially, it describes the form of virus transmission and general recommendations for its prevention. Subsequently, a detailed description is made of the biochemical aspects of SARS-CoV-2, its infectious cycle and the structure of protein S, which is involved in the process of entry of the virus into the cell. Finally, the methods and laboratory tests for the diagnosis of COVID-19 are described.


Asunto(s)
Humanos , Coronavirus , Células Epiteliales Alveolares , Tejido Parenquimatoso , Macrófagos
11.
Int. j. odontostomatol. (Print) ; 14(4): 501-507, dic. 2020. graf
Artículo en Español | LILACS | ID: biblio-1134527

RESUMEN

RESUMEN: Un nuevo coronavirus (SARS-CoV-2) ha sido reconocido como el agente etiológico de una misteriosa neumonía originada en Wuhan, China. La OMS ha nombrado a la nueva enfermedad como COVID-19 y, además, la ha declarado pandemia. Taxonómicamente, SARS-CoV-2 pertenece al género de los betacoronavirus junto con SARS-CoV y MERS-CoV. SARS-CoV-2 utiliza la enzima convertidora de la angiotensina 2 (ACE2) como el receptor objetivo para el ingreso en una célula huésped. La expresión de ACE2 en células de tejidos humanos podría indicar un potencial riesgo de reconocimiento por parte del virus y, por ende, ser susceptibles a la infección. Mediante algunas técnicas de laboratorio y de bioinformática, se ha visto una alta presencia de ACE2 en células epiteliales alveolares tipo II de pulmón y en enterocitos del intestino delgado. En la cavidad oral, se ha podido identificar la presencia de ACE2, principalmente, en células epiteliale s de glándulas salivales y células epiteliales de la lengua. Además, se ha reportado la manifestación de algunos síntomas, como sequedad bucal y ambligeustia, los que podrían estar relacionadas con una infección de SARS-CoV-2 en estos órganos. Sin embargo, son necesarios mayores estudios que evidencien esta situación.


ABSTRACT: A novel coronavirus (SARS-CoV-2) has been recognized as a etiologic agent of a mysterious pneumonia originating in Wuhan, China. WHO has named the new disease as COVID-19 and, in addition, has declared it a pandemic. Taxonomically, SARS-CoV-2 belongs to the betacoronavirus genus along with SARS-CoV and MERS-CoV. SARS-CoV-2 uses angiotensin-converting enzyme 2 (ACE2) as the target receptor for entry into a host cell. The expression of ACE2 in cells of human tissues could indicate a potential risk of recognition by the virus and, therefore, be susceptible to infection. Through some laboratory and bioinformatics techniques, high presence of ACE2 has been seen in type II alveolar epithelial cells of the lung and enterocytes of the small intestine. In oral cavity, mainly presence of ACE2 has been identified in epithelial cells of salivary glands and epithelial cells of tongue. In addition, manifestation of some symptoms, such as dry mouth and amblygeustia, have been reported, which could be related to a SARS-CoV-2 infection in these organs. However, further studies are needed to prove this situation.


Asunto(s)
Humanos , Inhibidores de la Enzima Convertidora de Angiotensina , Infecciones por Coronavirus/epidemiología , Peptidil-Dipeptidasa A/química , Betacoronavirus/química , Técnicas de Cultivo de Tejidos/métodos , Células Epiteliales Alveolares/citología , Células Epiteliales Alveolares/virología , Boca/virología
12.
Protein & Cell ; (12): 707-722, 2020.
Artículo en Inglés | WPRIM | ID: wpr-828750

RESUMEN

The 2019 novel coronavirus disease (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has occurred in China and around the world. SARS-CoV-2-infected patients with severe pneumonia rapidly develop acute respiratory distress syndrome (ARDS) and die of multiple organ failure. Despite advances in supportive care approaches, ARDS is still associated with high mortality and morbidity. Mesenchymal stem cell (MSC)-based therapy may be an potential alternative strategy for treating ARDS by targeting the various pathophysiological events of ARDS. By releasing a variety of paracrine factors and extracellular vesicles, MSC can exert anti-inflammatory, anti-apoptotic, anti-microbial, and pro-angiogenic effects, promote bacterial and alveolar fluid clearance, disrupt the pulmonary endothelial and epithelial cell damage, eventually avoiding the lung and distal organ injuries to rescue patients with ARDS. An increasing number of experimental animal studies and early clinical studies verify the safety and efficacy of MSC therapy in ARDS. Since low cell engraftment and survival in lung limit MSC therapeutic potentials, several strategies have been developed to enhance their engraftment in the lung and their intrinsic, therapeutic properties. Here, we provide a comprehensive review of the mechanisms and optimization of MSC therapy in ARDS and highlighted the potentials and possible barriers of MSC therapy for COVID-19 patients with ARDS.


Asunto(s)
Animales , Humanos , Traslado Adoptivo , Células Epiteliales Alveolares , Patología , Apoptosis , Betacoronavirus , Líquidos Corporales , Metabolismo , Linfocitos T CD4-Positivos , Alergia e Inmunología , Ensayos Clínicos como Asunto , Coinfección , Terapéutica , Infecciones por Coronavirus , Alergia e Inmunología , Modelos Animales de Enfermedad , Células Endoteliales , Patología , Oxigenación por Membrana Extracorpórea , Terapia Genética , Métodos , Vectores Genéticos , Usos Terapéuticos , Inmunidad Innata , Mediadores de Inflamación , Metabolismo , Pulmón , Patología , Trasplante de Células Madre Mesenquimatosas , Métodos , Células Madre Mesenquimatosas , Fisiología , Insuficiencia Multiorgánica , Pandemias , Neumonía Viral , Alergia e Inmunología , Síndrome de Dificultad Respiratoria , Alergia e Inmunología , Patología , Terapéutica , Investigación Biomédica Traslacional
13.
Protein & Cell ; (12): 707-722, 2020.
Artículo en Inglés | WPRIM | ID: wpr-828586

RESUMEN

The 2019 novel coronavirus disease (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has occurred in China and around the world. SARS-CoV-2-infected patients with severe pneumonia rapidly develop acute respiratory distress syndrome (ARDS) and die of multiple organ failure. Despite advances in supportive care approaches, ARDS is still associated with high mortality and morbidity. Mesenchymal stem cell (MSC)-based therapy may be an potential alternative strategy for treating ARDS by targeting the various pathophysiological events of ARDS. By releasing a variety of paracrine factors and extracellular vesicles, MSC can exert anti-inflammatory, anti-apoptotic, anti-microbial, and pro-angiogenic effects, promote bacterial and alveolar fluid clearance, disrupt the pulmonary endothelial and epithelial cell damage, eventually avoiding the lung and distal organ injuries to rescue patients with ARDS. An increasing number of experimental animal studies and early clinical studies verify the safety and efficacy of MSC therapy in ARDS. Since low cell engraftment and survival in lung limit MSC therapeutic potentials, several strategies have been developed to enhance their engraftment in the lung and their intrinsic, therapeutic properties. Here, we provide a comprehensive review of the mechanisms and optimization of MSC therapy in ARDS and highlighted the potentials and possible barriers of MSC therapy for COVID-19 patients with ARDS.


Asunto(s)
Animales , Humanos , Traslado Adoptivo , Células Epiteliales Alveolares , Patología , Apoptosis , Betacoronavirus , Líquidos Corporales , Metabolismo , Linfocitos T CD4-Positivos , Alergia e Inmunología , Ensayos Clínicos como Asunto , Coinfección , Terapéutica , Infecciones por Coronavirus , Alergia e Inmunología , Modelos Animales de Enfermedad , Células Endoteliales , Patología , Oxigenación por Membrana Extracorpórea , Terapia Genética , Métodos , Vectores Genéticos , Usos Terapéuticos , Inmunidad Innata , Mediadores de Inflamación , Metabolismo , Pulmón , Patología , Trasplante de Células Madre Mesenquimatosas , Métodos , Células Madre Mesenquimatosas , Fisiología , Insuficiencia Multiorgánica , Pandemias , Neumonía Viral , Alergia e Inmunología , Síndrome de Dificultad Respiratoria , Alergia e Inmunología , Patología , Terapéutica , Investigación Biomédica Traslacional
14.
Environmental Health and Preventive Medicine ; : 56-56, 2020.
Artículo en Inglés | WPRIM | ID: wpr-827259

RESUMEN

BACKGROUND@#We previously demonstrated that continuous exposure to nitrous acid gas (HONO) for 4 weeks, at a concentration of 3.6 parts per million (ppm), induced pulmonary emphysema-like alterations in guinea pigs. In addition, we found that HONO affected asthma symptoms, based on the measurement of respiratory function in rats exposed to 5.8 ppm HONO. This study aimed to investigate the dose-response effects of HONO exposure on the histopathological alterations in the respiratory tract of guinea pigs to determine the lowest observed adverse effect level (LOAEL) of HONO.@*METHODS@#We continuously exposed male Hartley guinea pigs (n = 5) to four different concentrations of HONO (0.0, 0.1, 0.4, and 1.7 ppm) for 4 weeks (24 h/day). We performed histopathological analysis by observing lung tissue samples. We examined samples from three guinea pigs in each group under a light microscope and measured the alveolar mean linear intercept (Lm) and the thickness of the bronchial smooth muscle layer. We further examined samples from two guinea pigs in each group under a scanning electron microscope (SEM) and a transmission electron microscope (TEM).@*RESULTS@#We observed the following dose-dependent changes: pulmonary emphysema-like alterations in the centriacinar regions of alveolar ducts, significant increase in Lm in the 1.7 ppm HONO-exposure group, tendency for hyperplasia and pseudostratification of bronchial epithelial cells, and extension of the bronchial epithelial cells and smooth muscle cells in the alveolar duct regions.@*CONCLUSIONS@#These histopathological findings suggest that the LOAEL of HONO is < 0.1 ppm.


Asunto(s)
Animales , Masculino , Células Epiteliales Alveolares , Bronquios , Relación Dosis-Respuesta a Droga , Enfisema , Células Epiteliales , Cobayas , Hiperplasia , Exposición por Inhalación , Pulmón , Patología , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Miocitos del Músculo Liso , Ácido Nitroso , Toxicidad
15.
Protein & Cell ; (12): 707-722, 2020.
Artículo en Inglés | WPRIM | ID: wpr-827023

RESUMEN

The 2019 novel coronavirus disease (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has occurred in China and around the world. SARS-CoV-2-infected patients with severe pneumonia rapidly develop acute respiratory distress syndrome (ARDS) and die of multiple organ failure. Despite advances in supportive care approaches, ARDS is still associated with high mortality and morbidity. Mesenchymal stem cell (MSC)-based therapy may be an potential alternative strategy for treating ARDS by targeting the various pathophysiological events of ARDS. By releasing a variety of paracrine factors and extracellular vesicles, MSC can exert anti-inflammatory, anti-apoptotic, anti-microbial, and pro-angiogenic effects, promote bacterial and alveolar fluid clearance, disrupt the pulmonary endothelial and epithelial cell damage, eventually avoiding the lung and distal organ injuries to rescue patients with ARDS. An increasing number of experimental animal studies and early clinical studies verify the safety and efficacy of MSC therapy in ARDS. Since low cell engraftment and survival in lung limit MSC therapeutic potentials, several strategies have been developed to enhance their engraftment in the lung and their intrinsic, therapeutic properties. Here, we provide a comprehensive review of the mechanisms and optimization of MSC therapy in ARDS and highlighted the potentials and possible barriers of MSC therapy for COVID-19 patients with ARDS.


Asunto(s)
Animales , Humanos , Traslado Adoptivo , Células Epiteliales Alveolares , Patología , Apoptosis , Betacoronavirus , Líquidos Corporales , Metabolismo , Linfocitos T CD4-Positivos , Alergia e Inmunología , Ensayos Clínicos como Asunto , Coinfección , Terapéutica , Infecciones por Coronavirus , Alergia e Inmunología , Modelos Animales de Enfermedad , Células Endoteliales , Patología , Oxigenación por Membrana Extracorpórea , Terapia Genética , Métodos , Vectores Genéticos , Usos Terapéuticos , Inmunidad Innata , Mediadores de Inflamación , Metabolismo , Pulmón , Patología , Trasplante de Células Madre Mesenquimatosas , Métodos , Células Madre Mesenquimatosas , Fisiología , Insuficiencia Multiorgánica , Pandemias , Neumonía Viral , Alergia e Inmunología , Síndrome de Dificultad Respiratoria , Alergia e Inmunología , Patología , Terapéutica , Investigación Biomédica Traslacional
16.
Rev. chil. enferm. respir ; 35(4): 261-263, dic. 2019. graf
Artículo en Español | LILACS | ID: biblio-1092702

RESUMEN

La fibrosis pulmonar idiopática (FPI) es una forma específica de neumonía intersticial idiopática, de tipo fibrosante crónica y progresiva, con patrón radiológico y/o histológico de neumonía intersticial usual (NIU). Su patogenia es compleja, el modelo más aceptado actualmente es basado en las células epiteliales alveolares, aberrantemente activadas que conducen a la proliferación de fibroblastos y su diferenciación a miofibroblastos que depositan matriz extracelular y destruyen irreversiblemente la arquitectura pulmonar. No existe un claro factor inicial que explique la activación y posterior mantención del mecanismo de la fibrosis. El factor de crecimiento transformante beta (TGF-β) liberado por las células epiteliales alveolares se ha implicado como unos de los principales conductores de la inducción y proliferación de fibroblastos alterados que persiste mucho tiempo después de la estimulación inicial, lo que explicaría en gran parte el comportamiento clínico progresivo y crónico.


Idiopathic pulmonary fibrosis (IPF) is a specific form of idiopathic interstitial pneumonia, of chronic and progressive fibrosing type, with radiological and / or histological pattern of usual interstitial pneumonia (UIP). Its pathogenesis is complex, the most accepted model currently is based on the fact that the alveolar epithelial cells, aberrantly activated, lead to the proliferation of fibroblasts and their differentiation to myofibroblasts that deposit extracellular matrix and irreversibly destroy the pulmonary architecture. There is no clear initial trigger that explains the activation and subsequent maintenance of the fibrosis mechanism. The transforming growth factor beta (TGF-β), released by the alveolar epithelial cells, has been implicated as one of the main drivers of the induction and proliferation of altered fibroblasts that persists long after the initial stimulation, which would largely explain progressive and chronic clinical behavior.


Asunto(s)
Humanos , Fibrosis Pulmonar Idiopática/etiología , Fibrosis Pulmonar Idiopática/fisiopatología , Fibrosis Pulmonar Idiopática/epidemiología , Factores de Riesgo , Factor de Crecimiento Transformador beta , Matriz Extracelular , Células Epiteliales Alveolares
17.
Acta Physiologica Sinica ; (6): 575-580, 2019.
Artículo en Chino | WPRIM | ID: wpr-777154

RESUMEN

The aim of the present study was to investigate the effect of salidroside (Sal) on inflammatory activation induced by lipopolysaccharide (LPS) in the co-culture of rat alveolar macrophages (AM) NR 8383 and type II alveolar epithelial cells (AEC II) RLE-6TN. CCK-8 colorimetric method was used to detect cell proliferation percentage. The enzyme-linked immunosorbent assay (ELISA) was used to determine the content of tumor necrosis factor alpha (TNF-α), macrophage inflammatory protein-2 (MIP-2) and interleukin-10 (IL-10) in the supernatant. Western blot was used to examine the expression levels of phosphorylated AKT (p-AKT) and total AKT protein. The results showed that pretreatment of RLE-6TN cells or co-culture of RLE-6TN and NR 8383 cells with 32 and 128 µg/mL Sal for 1 h, followed by continuous culture for 24 h, significantly increased the cell proliferation (P < 0.05). Compared with control group, 32 and 128 µg/mL Sal pretreatment significantly increased the ratio of p-AKT/AKT in RLE-6TN cells (P < 0.05). Pretreatment of 32 µg/mL Sal not only inhibited the secretion of TNF-α and MIP-2 by NR 8383 cells induced by LPS (P < 0.05), but also enhanced the inhibitory effect of RLE-6TN and NR 8383 cells co-culture on the secretion of TNF-α and MIP-2 by NR 8383 cells induced by LPS (P < 0.05). In addition, 32 µg/mL Sal pretreatment promoted LPS-induced IL-10 secretion by NR 8383 cells (P < 0.05), and enhanced the promoting effect of co-culture of RLE-6TN and NR 8383 cells on the IL-10 secretion by LPS-induced NR 8383 cells (P < 0.05). In conclusion, Sal may directly inhibit LPS-induced inflammatory activation of AM (NR 8383), promote the proliferation of AEC II (RLE-6TN) through PI3K/AKT signaling pathway, and enhance the regulatory effect of AEC II on LPS-induced inflammatory activation of AM.


Asunto(s)
Animales , Ratas , Células Epiteliales Alveolares , Metabolismo , Línea Celular , Quimiocina CXCL2 , Metabolismo , Técnicas de Cocultivo , Glucósidos , Farmacología , Interleucina-10 , Metabolismo , Lipopolisacáridos , Macrófagos Alveolares , Metabolismo , Fenoles , Farmacología , Fosfatidilinositol 3-Quinasas , Metabolismo , Proteínas Proto-Oncogénicas c-akt , Metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa , Metabolismo
18.
Braz. j. med. biol. res ; 51(2): e6950, 2018. tab, graf
Artículo en Inglés | LILACS | ID: biblio-889028

RESUMEN

Alveolar epithelia play an essential role in maintaining the integrity and homeostasis of lungs, in which alveolar epithelial type II cells (AECII) are a cell type with stem cell potential for epithelial injury repair and regeneration. However, mechanisms behind the physiological and pathological roles of alveolar epithelia in human lungs remain largely unknown, partially owing to the difficulty of isolation and culture of primary human AECII cells. In the present study, we aimed to characterize alveolar epithelia generated from A549 lung adenocarcinoma cells that were cultured in an air-liquid interface (ALI) state. Morphological analysis demonstrated that A549 cells could reconstitute epithelial layers in ALI cultures as evaluated by histochemistry staining and electronic microscopy. Immunofluorescent staining further revealed an expression of alveolar epithelial type I cell (AECI) markers aquaporin-5 protein (AQP-5), and AECII cell marker surfactant protein C (SPC) in subpopulations of ALI cultured cells. Importantly, molecular analysis further revealed the expression of AQP-5, SPC, thyroid transcription factor-1, zonula occludens-1 and Mucin 5B in A549 ALI cultures as determined by both immunoblotting and quantitative RT-PCR assay. These results suggest that the ALI culture of A549 cells can partially mimic the property of alveolar epithelia, which may be a feasible and alternative model for investigating roles and mechanisms of alveolar epithelia in vitro.


Asunto(s)
Humanos , Medios de Cultivo Condicionados , Técnicas de Cultivo de Célula/métodos , Células Epiteliales Alveolares/fisiología , Células A549/fisiología , Valores de Referencia , Factores de Tiempo , Microscopía Electrónica de Rastreo , Immunoblotting , Recuento de Células , Reproducibilidad de los Resultados , Análisis de Varianza , Proteína C Asociada a Surfactante Pulmonar/análisis , Acuaporina 5/análisis , Mucina 5B/análisis , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína de la Zonula Occludens-1/análisis , Factor Nuclear Tiroideo 1/análisis
19.
Rev. bras. anestesiol ; 67(6): 600-606, Nov.-Dec. 2017. graf
Artículo en Inglés | LILACS | ID: biblio-897789

RESUMEN

Abstract Background and objectives Dexmedetomidine (DEX) has demonstrated the preconditioning effect and shown protective effects against organize injury. In this study, using A549 (human alveolar epithelial cell) cell lines, we investigated whether DEX preconditioning protected against acute lung injury (ALI) in vitro. Methods A549 were randomly divided into four groups (n = 5): control group, DEX group, lipopolysaccharides (LPS) group, and D-LPS (DEX + LPS) group. Phosphate buffer saline (PBS) or DEX were administered. After 2 h preconditioning, the medium was refreshed and the cells were challenged with LPS for 24 h on the LPS and D-LPS group. Then the malondialdehyde (MDA), superoxide dismutase (SOD), Bcl-2, Bax, caspase-3 and the cytochrome c in the A549 were tested. The apoptosis was also evaluated in the cells. Results Compare with LPS group, DEX preconditioning reduced the apoptosis (26.43% ± 1.05% vs. 33.58% ± 1.16%, p < 0.05) in the A549, which is correlated with decreased MDA (12.84 ± 1.05 vs. 19.16 ± 1.89 nmoL.mg-1 protein, p < 0.05) and increased SOD activity (30.28 ± 2.38 vs. 20.86 ± 2.19 U.mg-1 protein, p < 0.05). DEX preconditioning also increased the Bcl-2 level (0.53 ± 0.03 vs. 0.32 ± 0.04, p < 0.05) and decreased the level of Bax (0.49 ± 0.04 vs. 0.65 ± 0.04, p < 0.05), caspase-3 (0.54 ± 0.04 vs. 0.76 ± 0.04, p < 0.05) and cytochrome c. Conclusion DEX preconditioning has a protective effect against ALI in vitro. The potential mechanisms involved are the inhibition of cell death and improvement of antioxidation.


Resumo Justificativa e objetivos Dexmedetomidina (DEX) demonstrou ter efeito pré-condicionante e também efeitos protetores contra lesão organizada. Neste estudo, com células A549 (células epiteliais alveolares humanas), investigamos se o pré-condicionamento com DEX proporcionaria proteção contra lesão pulmonar aguda (LPA) in vitro. Métodos Células A549 foram aleatoriamente distribuídas em quatro grupos (n = 5): controle, DEX, lipopolissacarídeos (LPS) e D-LPS (DEX + LPS). Administramos solução de PBS (tampão fosfato-alcalino) ou DEX. Após 2 h de pré-condicionamento, o meio foi renovado e as células desafiadas com LPS por 24 h nos grupos LPS e D-LPS. Em seguida, malondialdeído (MDA), superóxido dismutase (SOD), Bcl-2, Bax, caspase-3 e em A549 foram testados. Apoptose também foi avaliada nas células. Resultados Em comparação com o grupo LPS, o pré-condicionamento com DEX reduziu a apoptose (26,43% ± 1,05% vs. 33,58% ± 1,16%, p < 0,05) em células A549, o que está correlacionado com a diminuição de MDA (12,84 ± 1,05 vs. 19,16 ± 1,89 nmol.mg-1 de proteína, p < 0,05) e aumento da atividade de SOD (30,28 ± 2,38 vs. 20,86 ± 2,19 U.mg-1 de proteína, p < 0,05). O pré-condicionamento com DEX também aumentou o nível de Bcl-2 (0,53 ± 0,03 vs. 0,32 ± 0,04, p < 0,05) e diminuiu o nível de Bax (0,49 ± 0,04 vs. 0,65 ± 0,04, p < 0,05), caspase-3 (0,54 ± 0,04 vs. 0,76 ± 0,04, p < 0,05) e citocromo c. Conclusão O pré-condicionamento com DEX tem efeito protetor contra LPA in vitro. Os potenciais mecanismos envolvidos são inibição da morte celular e melhoria da antioxidação.


Asunto(s)
Humanos , Lipopolisacáridos/efectos adversos , Dexmedetomidina/farmacología , Células Epiteliales Alveolares/efectos de los fármacos , Hipnóticos y Sedantes/farmacología , Distribución Aleatoria , Células Cultivadas , Lipopolisacáridos/antagonistas & inhibidores
20.
Arq. bras. med. vet. zootec. (Online) ; 69(3): 593-596, jun. 2017. ilus
Artículo en Portugués | LILACS, VETINDEX | ID: biblio-846892

RESUMEN

As doenças pulmonares intersticiais constituem um grupo de doenças difusas do parênquima pulmonar, no qual a fibrose pulmonar intersticial está incluída. Histologicamente, esta se caracteriza por hiperplasia de pneumócitos tipo II, hiperplasia ou hipertrofia de músculo liso e fibrose. Embora a patogenia da fibrose pulmonar intersticial não esteja bem elucidada, devido às semelhanças microscópicas encontradas nos pneumócitos tipo II em felinos e na forma familiar da doença em humanos, acredita-se que haja caráter genético para o seu desenvolvimento. Os sinais clínicos frequentemente relatados incluem desconforto respiratório, cianose, letargia e perda de peso. Devido ao caráter progressivo e à ausência de tratamento específico, a doença apresenta prognóstico desfavorável. Foi atendida uma gata de 12 anos de idade, com histórico de dispneia há 20 dias. Ao exame clínico, o animal apresentou dispneia expiratória restritiva, crepitação à ausculta torácica e foi visualizado padrão intersticial ao exame radiográfico do tórax. A paciente foi submetida à punção com agulha fina de tecido pulmonar e veio a óbito algumas horas após o procedimento, apresentando insuficiência respiratória aguda. No exame histológico do tecido pulmonar, foi verificada a ocorrência de fibrose pulmonar idiopática. O objetivo do presente trabalho é relatar um caso de dispneia expiratória restritiva em um felino doméstico devido à fibrose pulmonar idiopática, já que, segundo o conhecimento dos autores, não há nenhum relato da ocorrência da doença no país.(AU)


Interstitial lung diseases are a group of diffuse parenchymal lung diseases in which interstitial lung fibrosis is included. Histologically, it is characterized by type II pneumocyte hyperplasia, hypertrophy or hyperplasia of smooth tissue and fibrosis. Although the pathogenesis of interstitial lung fibrosis has not been elucidated, due to the microscopic similarities found in type II pneumocytes in cats and familial form of the disease in humans, it is believed that there is a genetic trait for development. The frequently reported clinical signs include respiratory distress, cyanosis, lethargy, and weight loss. Due to the progressive nature and the absence of specific treatment, the disease has a poor prognosis. A 12-year-old cat with dyspnea for 20 days was assisted. The animal underwent fine needle aspiration of lung tissue and died few hours after the procedure, with acute respiratory failure. Upon histological examination of lung tissue, the occurrence of idiopathic pulmonary fibrosis was found. The aim of this study is to report a case of restrictive expiratory dyspnea in a domestic feline due to idiopathic pulmonary fibrosis, because, according to our knowledge, there is no report on the occurrence of the disease in our country.(AU)


Asunto(s)
Animales , Gatos , Disnea/veterinaria , Fibrosis Pulmonar Idiopática/veterinaria , Células Epiteliales Alveolares , Enfermedades Pulmonares Intersticiales/veterinaria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA