Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 268
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Ecotoxicol Environ Saf ; 277: 116357, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38677073

RESUMO

Polystyrene microplastics (PS-MPs) are new types of environmental pollutant that have garnered significant attention in recent years since they were found to cause damage to the human respiratory system when they are inhaled. The pulmonary fibrosis is one of the serious consequences of PS-MPs inhalation. However, the impact and underlying mechanisms of PS-MPs on pulmonary fibrosis are not clear. In this study, we studied the potential lung toxicity and PS-MPs-developed pulmonary fibrosis by long-term intranasal inhalation of PS-MPs. The results showed that after exposing to the PS-MPs, the lungs of model mouse had different levels of damage and fibrosis. Meanwhile, exposing to the PS-MPs resulted in a markedly decrease in glutathione (GSH), an increase in malondialdehyde (MDA), and iron overload in the lung tissue of mice and alveolar epithelial cells (AECs). These findings suggested the occurrence of PS-MP-induced ferroptosis. Inhibitor of ferroptosis (Fer-1) had alleviated the PS-MPs-induced ferroptosis. Mechanically, PS-MPs triggered cell ferroptosis and promoted the development of pulmonary fibrosis via activating the cGAS/STING signaling pathway. Inhibition of cGAS/STING with G150/H151 attenuated pulmonary fibrosis after PS-MPs exposure. Together, these data provided novel mechanistic insights of PS-MPs-induced pulmonary fibrosis and a potential therapeutic paradigm.


Assuntos
Células Epiteliais Alveolares , Ferroptose , Proteínas de Membrana , Microplásticos , Poliestirenos , Fibrose Pulmonar , Transdução de Sinais , Ferroptose/efeitos dos fármacos , Animais , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/patologia , Poliestirenos/toxicidade , Camundongos , Transdução de Sinais/efeitos dos fármacos , Microplásticos/toxicidade , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/patologia , Proteínas de Membrana/metabolismo , Masculino , Camundongos Endogâmicos C57BL
2.
Environ Toxicol ; 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38899512

RESUMO

Despite recent advances in treatment, non-small cell lung cancer (NSCLC) continues to have a high mortality rate. Currently, NSCLC pathogenesis requires further investigation, and therapeutic drugs are still under development. Homologous recombination repair (HRR) repairs severe DNA double-strand breaks. Homologous recombination repair deficiency (HRD) occurs when HRR is impaired and causes irreparable double-strand DNA damage, leading to genomic instability and increasing the risk of cancer development. Poly(ADP-ribose) polymerase (PARP) inhibitors can effectively treat HRD-positive tumors. Extracellular heat shock protein 90α (eHSP90α) is highly expressed in hypoxic environments and inhibits apoptosis, thereby increasing cellular tolerance. Here, we investigated the relationship between eHSP90α and HRR in NSCLC. DNA damage models were established in NSCLC cell lines (A549 and H1299). The activation of DNA damage and HRR markers, apoptosis, proliferation, and migration were investigated. In vivo tumor models were established using BALB/c nude mice and A549 cells. We found that human recombinant HSP90α stimulation further activated HRR and reduced DNA damage extent; however, eHSP90α monoclonal antibody, 1G6-D7, effectively inhibited HRR. HRR inhibition and increased apoptosis were observed after LRP1 knockdown; this effect could not be reversed with hrHSP90α addition. The combined use of 1G6-D7 and olaparib caused significant apoptosis and HRR inhibition in vitro and demonstrated promising anti-tumor effects in vivo. Extracellular HSP90α may be involved in HRR in NSCLC through LRP1. The combined use of 1G6-D7 and PARP inhibitors may exert anti-tumor effects by inhibiting DNA repair and further inducing apoptosis of NSCLC cells.

3.
J Transl Med ; 21(1): 828, 2023 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-37978543

RESUMO

BACKGROUND: The tumor microenvironment plays a key role in non-small cell lung cancer (NSCLC) development and also influences the effective response to immunotherapy. The pro-inflammatory factor interleukin-17A mediates important immune responses in the tumor microenvironment. In this study, the potential role and mechanisms of IL-17A in NSCLC were investigated. METHODS: We detected IL-17A by immunohistochemistry (IHC) in 39 NSCLC patients. Its expression was correlated with the programmed cell death-ligand1 (PD-L1). IL-17A knockdown and overexpression in A549 and SPC-A-1 cell models were constructed. The function of IL-17A was examined in vitro by wound healing, migration, invasion, plate colony formation and T cell killing assay. Western blot analysis, immunofluorescence assay and IHC were performed to investigate the regulation effects of IL-17A on autophagy in A549 and SPC-A-1. The effect of IL-17A on ROS/Nrf2/p62 signaling pathway was detected. Subcutaneous tumor models were established to examine the tumor-promoting effect of IL-17A in vivo and its effect on immunotherapy. RESULTS: We found a prevalent expression of IL-17A in NSCLC tumor tissues and it was positively correlated with PD-L1 expression (r = 0.6121, p < 0.0001). In vitro, IL-17A promotes lung cancer cell migration, invasion and colony formation ability. Moreover, IL-17A upregulated N-cadherin, Twist, and Snail, and downregulated E-cadherin in NSCLC cells. IL-17A enhanced cell survival in the T cell killing assay. Mechanistically, IL-17A induced ROS production and increased Nrf2 and p62 expression, thereby inhibiting autophagy and reducing PD-L1 degradation. In vivo experiments, anti-IL-17A monoclonal antibody alone slowed the growth of subcutaneous tumors in mice. When combined with anti-PD-L1 monoclonal antibody, tumor tissue expression of PD-L1 was reduced and the therapeutic effect was diminished. CONCLUSION: We found that IL-17A promoted NSCLC progression and inhibited autophagy through the ROS/Nrf2/p62 pathway leading to increased PD-L1 expression in cancer cells. Modulation of IL-17A may affect the therapeutic efficacy of immunotherapy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Animais , Camundongos , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/patologia , Interleucina-17/metabolismo , Antígeno B7-H1/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Espécies Reativas de Oxigênio , Transformação Celular Neoplásica , Carcinogênese , Anticorpos Monoclonais/uso terapêutico , Apoptose , Microambiente Tumoral
4.
Respir Res ; 24(1): 8, 2023 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-36627645

RESUMO

BACKGROUND: Lung fibroblast activation is associated with airway remodeling during asthma progression. Stearoyl-CoA desaturase 1 (SCD1) plays an important role in the response of fibroblasts to growth factors. This study aimed to explore the effects of SCD1 on fibroblast activation induced by transforming growth factor-ß1 (TGF-ß1) and the role of the phosphatidylinositol-3-kinase-AKT serine-threonine protein kinase-mechanistic target of rapamycin (PI3K-Akt-mTOR) pathway on the regulation of SCD1 expression in airway remodeling. METHODS: Female C57BL/6 mice were sensitized and challenged with house dust mites to generate a chronic asthma model. The inhibitor of SCD1 was injected i.g. before each challenge. The airway hyper-responsiveness to methacholine was evaluated, and airway remodeling and airway inflammation were assessed by histology. The effects of SCD1 on fibroblast activation were evaluated in vitro using an SCD1 inhibitor and oleic acid and via the knockdown of SCD1. The involvement of the PI3K-Akt-mTOR-sterol regulatory element-binding protein 1 (SREBP1) pathway in lung fibroblasts was investigated using relevant inhibitors. RESULTS: The expression of SCD1 was increased in fibroblasts exposed to TGF-ß1. The inhibition of SCD1 markedly ameliorated airway remodeling and lung fibroblast activation in peripheral airways. The knockdown or inhibition of SCD1 resulted in significantly reduced extracellular matrix production in TGF-ß1-treated fibroblasts, but this effect was reversed by the addition of exogenous oleic acid. The PI3K-Akt-mTOR-SREBP1 pathway was found to be involved in the regulation of SCD1 expression and lung fibroblast activation. CONCLUSIONS: The data obtained in this study indicate that SCD1 expression contributes to fibroblast activation and airway remodeling and that the inhibition of SCD1 may be a therapeutic strategy for airway remodeling in asthma.


Assuntos
Asma , Proteínas Proto-Oncogênicas c-akt , Animais , Camundongos , Feminino , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Ácido Oleico/farmacologia , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/farmacologia , Remodelação das Vias Aéreas , Camundongos Endogâmicos C57BL , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Pulmão/metabolismo , Asma/patologia , Fibroblastos/metabolismo , Sirolimo/farmacologia , Estearoil-CoA Dessaturase/genética , Estearoil-CoA Dessaturase/metabolismo
5.
FASEB J ; 36(6): e22359, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35621121

RESUMO

Asthma is a disease characterized by airway epithelial barrier destruction, chronic airway inflammation, and airway remodeling. Repeated damage to airway epithelial cells by allergens in the environment plays an important role in the pathophysiology of asthma. Ferroptosis is a novel form of regulated cell death mediated by lipid peroxidation in association with free iron-mediated Fenton reactions. In this study, we explored the contribution of ferroptosis to house dust mite (HDM)-induced asthma models. Our in vivo and in vitro models showed labile iron accumulation and enhanced lipid peroxidation with concomitant nonapoptotic cell death upon HDM exposure. Treatment with ferroptosis inhibitors deferoxamine (DFO) and ferrostatin-1 (Fer-1) illuminated the role of ferroptosis and related damage-associated molecular patterns in HDM-treated airway epithelial cells. Furthermore, DFO and Fer-1 reduced HDM-induced airway inflammation in model mice. Mechanistically, NCOA4-mediated ferritin-selective autophagy (ferritinophagy) was initiated during ferritin degradation in response to HDM exposure. Together, these data suggest that ferroptosis plays an important role in HDM-induced asthma and that ferroptosis may be a potential treatment target for HDM-induced asthma.


Assuntos
Asma , Ferroptose , Animais , Células Epiteliais/metabolismo , Ferritinas/metabolismo , Inflamação , Ferro/metabolismo , Camundongos , Pyroglyphidae
6.
FASEB J ; 36(8): e22475, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35899478

RESUMO

Recent findings suggest that extracellular heat shock protein 90α (eHSP90α) promotes pulmonary fibrosis, but the underlying mechanisms are not well understood. Aging, especially cellular senescence, is a critical risk factor for idiopathic pulmonary fibrosis (IPF). Here, we aim to investigate the role of eHSP90α on cellular senescence in IPF. Our results found that eHSP90α was upregulated in bleomycin (BLM)-induced mice, which correlated with the expression of senescence markers. This increase in eHSP90α mediated fibroblast senescence and facilitated mitochondrial dysfunction. eHSP90α activated TGF-ß signaling through the phosphorylation of the SMAD complex. The SMAD complex binding to p53 and p21 promoters triggered their transcription. In vivo, the blockade of eHSP90α with 1G6-D7, a specific eHSP90α antibody, in old mice attenuated the BLM-induced lung fibrosis. Our findings elucidate a crucial mechanism underlying eHSP90α-induced cellular senescence, providing a framework for aging-related fibrosis interventions.


Assuntos
Bleomicina , Fibrose Pulmonar Idiopática , Animais , Bleomicina/toxicidade , Senescência Celular , Fibroblastos/metabolismo , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/metabolismo , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fator de Crescimento Transformador beta/metabolismo
7.
Am J Respir Cell Mol Biol ; 66(6): 648-660, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35358396

RESUMO

Thymic stromal lymphopoietin presents in two distinct isoforms: short-form (sfTSLP) and long-form (lfTSLP). lfTSLP promotes inflammation, whereas sfTSLP inhibits inflammation, in allergic asthma. However, little is known about the regulation of lfTSLP and sfTSLP during allergic attack in the asthma airway epithelium. Here, we report that small ubiquitin-like modifier (SUMOylation) was enhanced in house dust mite-induced allergic asthma airway epithelium. Inhibition of SUMOylation significantly alleviated airway T-helper cell type 2 inflammation and lfTSLP expression. Mechanistically, chromobox 4 (CBX4), a SUMOylation E3 ligase, enhanced lfTSLP mRNA translation, but not sfTSLP, through the RNA-binding protein muscle excess (MEX)-3B. MEX-3B promoted lfTSLP translation by binding the lfTSLP mRNA through its K homology domains. Furthermore, CBX4 regulated MEX-3B transcription in human bronchial epithelial cells through enhancing SUMOylation concentrations of the transcription factor TFII-I. In conclusion, we demonstrate an important mechanism whereby CBX4 promotes MEX-3B transcription through enhancing TFII-I SUMOylation and MEX-3B enhances the expression of lfTSLP through binding to the lfTSLP mRNA and promoting its translation. Our findings uncover a novel target of CBX4 for therapeutic agents for lfTSLP-mediated asthma.


Assuntos
Asma , Citocinas , Ligases , Proteínas do Grupo Polycomb , Pyroglyphidae , Sumoilação , Animais , Asma/imunologia , Asma/metabolismo , Citocinas/metabolismo , Humanos , Inflamação , Ligases/metabolismo , Proteínas do Grupo Polycomb/metabolismo , Pyroglyphidae/imunologia , RNA Mensageiro/metabolismo , Linfopoietina do Estroma do Timo
8.
J Cell Mol Med ; 26(19): 4974-4985, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36128650

RESUMO

Orai family are a calcium channel of cell membrane extracellular Ca2+ influx which participates in tissue fibrosis. But the roles of Orai3 have less attention on the mechanism of regulating lung fibrosis. In this study, we found that Orai3 expression was increased significantly in BLM-induced lung fibrosis. The knockdown of Orai3 decreased TGF-ß1-induced fibroblast proliferation, ECM production, activation of NFAT1 and Calpain/ERK signal pathway and glycolysis levels. Orai3 interacting with Orai1 was increased in BLM-induced lung fibrosis and TGF-ß1-induced fibroblast, while the Stim1 interacting with Orai1 and SOCE activity was suppressed, leading in a high and stable extracellular Ca2+ influx. Furthermore, the over-expression of Orai3 did not enhance Orai3 interacting with Orai1 under TGF-ß1 free fibroblast. And then, the deeper mechanism of TGF-ß1-induced increased SEPTIN4 promoted Orai3 interacting with Orai1. Our results indicated that Orai3 could be one of the therapy targets for PF in which remodels Orai channel, suppresses SOCE activity and activated fibroblast to alleviate fibrosis progress.


Assuntos
Fibrose Pulmonar , Cálcio/metabolismo , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Sinalização do Cálcio , Calpaína/metabolismo , Fibroblastos/metabolismo , Humanos , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Fibrose Pulmonar/genética , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/metabolismo
9.
Respir Res ; 23(1): 75, 2022 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-35351157

RESUMO

BACKGROUND: Up-regulation of aerobic glycolysis has been reported as a characterization of asthma and facilitates airway inflammation. We has been previously reported that short isoform thymic stromal lymphopoietin (sTSLP) could reduce inflammation in asthmatic airway epithelial cells. Here we wanted to investigate whether the inhibition of sTSLP on asthma is related to aerobic glycolysis. METHODS: Asthmatic model was established in challenging Male BALB/c mice and 16-HBE (human bronchial epithelial) cell line with house dust mite (HDM). Indicators of glycolysis were assessed to measure whether involve in sTSLP regulating airway epithelial cells inflammation in asthmatic model in vivo and in vitro. RESULTS: sTSLP decreased inflammation of asthmatic airway and aerobic glycolysis in mice. HDM or long isoform thymic stromal lymphopoietin (lTSLP) promoted HIF-1α expression and aerobic glycolysis by miR-223 to target and inhibit VHL (von Hippel-Lindau) expression 16-HBE. Inhibition of aerobic glycolysis restrained HDM- and lTSLP-induced inflammatory cytokines production. sTSLP along had almost no potential to alter aerobic glycolysis of 16-HBE. But sTSLP decreased LDHA (lactate dehydrogenase A) and LD (Lactic acid) levels in BALF, and HIF-1α and LDHA protein levels in airway epithelial cells of asthma mice model. lTSLP and sTSLP both induced formation of TSLPR and IL-7R receptor complex, and lTSLP obviously facilitated phosphorylation of JAK1, JAK2 and STAT5, while sTSLP induced a little phosphorylation of JAK1 and STAT5. CONCLUSION: We identified a novel mechanism that lTSLP could promote inflammatory cytokines production by miR-223/VHL/HIF-1α pathway to upregulate aerobic glycolysis in airway epithelial cells in asthma. This pathway is suppressed by sTSLP through occupying binding site of lTSLP in TSLPR and IL-7R receptor complex.


Assuntos
Asma , Citocinas , Animais , Asma/metabolismo , Citocinas/metabolismo , Epitélio/metabolismo , Glicólise , Humanos , Inflamação/metabolismo , Masculino , Camundongos , Isoformas de Proteínas , Linfopoietina do Estroma do Timo
10.
BMC Pulm Med ; 22(1): 61, 2022 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-35148729

RESUMO

BACKGROUND: Exposure to toluene diisocyanate (TDI) is a significant pathogenic factor for asthma. We previously reported that the receptor for advanced glycation end products (RAGE) plays a key role in TDI-induced asthma. Histone deacetylase (HDAC) has been reported to be important in asthmatic pathogenesis. However, its effect on TDI-induced asthma is not known. The aim of this study was to determine the role of RAGE and HDAC in regulating airway inflammation using a TDI-induced murine asthma model. METHODS: BALB/c mice were sensitized and challenged with TDI to establish an asthma model. FPS-ZM1 (RAGE inhibitor), JNJ-26482585 and romidepsin (HDAC inhibitors) were administered intraperitoneally before each challenge. In vitro, the human bronchial epithelial cell line 16HBE was stimulated with TDI-human serum albumin (TDI-HSA). RAGE knockdown cells were constructed and evaluated, and MK2006 (AKT inhibitor) was also used in the experiments. RESULTS: In TDI-induced asthmatic mice, the expression of RAGE, HDAC1, and p-AKT/t-AKT was upregulated, and these expressions were attenuated by FPS-ZM1. Airway reactivity, Th2 cytokine levels in lymph supernatant, IgE, airway inflammation, and goblet cell metaplasia were significantly increased in the TDI-induced asthmatic mice. These increases were suppressed by JNJ-26482585 and romidepsin. In addition, JNJ-26482585 and romidepsin ameliorated the redistribution of E-cadherin and ß-catenin in TDI-induced asthma. In TDI-HSA-stimulated 16HBE cells, knockdown of RAGE attenuated the upregulation of HDAC1 and phospho-AKT (p-AKT). Treatment with the AKT inhibitor MK2006 suppressed TDI-induced HDAC1 expression. CONCLUSIONS: These findings indicate that RAGE modulates HDAC1 expression via the PI3K/AKT pathway, and that inhibition of HDAC prevents TDI-induced airway inflammation.


Assuntos
Asma/prevenção & controle , Histona Desacetilase 1/metabolismo , Inflamação/prevenção & controle , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Asma/induzido quimicamente , Benzamidas/farmacologia , Linhagem Celular , Citocinas/metabolismo , Depsipeptídeos/farmacologia , Modelos Animais de Doenças , Histona Desacetilase 1/antagonistas & inibidores , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fosfatidilinositol 3-Quinases/metabolismo , Receptor para Produtos Finais de Glicação Avançada/antagonistas & inibidores , Tolueno 2,4-Di-Isocianato/toxicidade
11.
BMC Pulm Med ; 22(1): 261, 2022 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-35778740

RESUMO

PURPOSE: While asthma comorbidities are associated with higher health care utilisation, lower quality of life and poorer asthma control, the impact of asthma comorbidities on hospitalisation for asthma exacerbation (H-AX) remains less recognised. We aim to analyse the impact of asthma comorbidities on H-AX. METHODS: Based on a national survey on asthma control and disease perception (CARN 2015 study), we analysed the impact of comorbidities on annual incidence and frequency of H-AX in China. Information on demographic characteristics, asthma comorbidities and annual incidence and frequency of H-AX were presented in this study. RESULTS: Among 3875 ambulatory asthma patients, 75.9% (2941/3875) had comorbidities, and 26.4% (1017/3858) experienced H-AX during past year. After adjusting for confounding factors such as demographic data, smoking status and asthma control, COPD [OR = 2.189, 95% CI (1.673, 2.863)] and coronary heart disease [OR = 1.387, 95% CI (1.032, 1.864)] were associated with higher annual incidence, while allergic rhinitis [OR = 0.692, 95% CI (0.588, 0.815)] was associated with lower annual incidence, of H-AX. In terms of frequency, allergic rhinitis [OR = 1.630, 95% CI (1.214, 2.187)], COPD [OR = 1.472, 95% CI (1.021, 2.122)] and anxiety [OR = 2.609, 95% CI (1.051, 6.477)] showed statistically significant correlation with frequent H-AX. CONCLUSIONS: COPD and coronary heart disease were associated with higher annual incidence, while allergic rhinitis was associated with lower annual incidence of H-AX. Allergic rhinitis, COPD and anxiety were associated with frequent H-AX. Comorbidities may have an important role in the risk and frequency of annual hospitalisations due to asthma exacerbation. The goal of asthma control should rely on a multi-disciplinary treatment protocol.


Assuntos
Asma , Doença Pulmonar Obstrutiva Crônica , Rinite Alérgica , Asma/complicações , Asma/epidemiologia , Hospitalização , Humanos , Incidência , Doença Pulmonar Obstrutiva Crônica/complicações , Doença Pulmonar Obstrutiva Crônica/epidemiologia , Qualidade de Vida , Rinite Alérgica/epidemiologia
12.
BMC Pulm Med ; 19(1): 218, 2019 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-31747880

RESUMO

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


Assuntos
Asma/fisiopatologia , Brônquios/efeitos dos fármacos , Cromonas/farmacologia , Células Epiteliais/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/metabolismo , Morfolinas/farmacologia , Animais , Asma/tratamento farmacológico , Brônquios/enzimologia , Brônquios/imunologia , Caderinas/metabolismo , Linhagem Celular , Citocinas/metabolismo , Impedância Elétrica , Células Epiteliais/enzimologia , Células Epiteliais/imunologia , Técnicas de Silenciamento de Genes , Proteínas de Choque Térmico HSP90/genética , Humanos , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fosforilação , Pyroglyphidae/imunologia , Mucosa Respiratória/metabolismo
13.
Carcinogenesis ; 39(10): 1216-1221, 2018 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-29939218

RESUMO

Lung cancer is a common cancer in human and has presented significant genetic predisposition. Previous genome-wide association study observed that rs401681 within CLPTM1L (CLPTM1 like) was significantly associated with lung cancer. By analyzing 1000 genomes data for East Asian, we identified only one SNP in nearby region, rs402710, in high linkage disequilibrium with rs401681, which was also associated with lung cancer. However, the real causal SNP and mechanism for the association were still not clear. The following plasmid construction, mutagenesis, transient transfection and luciferase reading indicated that both SNPs could regulate gene expression in lung/bronchial epithelium Beas-2B cell line. By chromosome conformation capture, it was identified that the segment containing these two SNPs could interact with TERT (telomerase reverse transcriptase) promoter, thus indicating that these SNPs confer lung cancer risk by regulating TERT expression instead of CLPTM1L. Through chromatin immunoprecipitation, the transcript factors HNF4A (hepatocyte nuclear factor 4 alpha) and MAF1 (MAF1 homolog, negative regulator of RNA polymerase III) were recognized for the regions spanning rs401681 and rs402710, respectively. Our results uncovered a complete link between these two SNPs and lung cancer.


Assuntos
Povo Asiático/genética , Neoplasias Pulmonares/genética , Proteínas de Membrana/genética , Proteínas de Neoplasias/genética , Telomerase/genética , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina/métodos , Regulação Neoplásica da Expressão Gênica/genética , Predisposição Genética para Doença , Genótipo , Humanos , Neoplasias Pulmonares/etnologia , Polimorfismo de Nucleotídeo Único
14.
Biochem Biophys Res Commun ; 496(1): 238-243, 2018 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-29309789

RESUMO

To build a microfluidic device with various morphological features of the tumor vasculature for study of the effects of tumor vascular structures on the flow field and tumor cellular flow behaviors. The designed microfluidic device was able to approximatively simulate the in vivo structures of tumor vessels and the flow within it. In this models, the influences of the angle of bifurcation, the number of branches, and the narrow channels on the flow field and the influence of vorticity on the retention of HepG2 cells were significant. Additionally, shear stress below physiological conditions of blood circulation has considerable effect on the formation of the lumen-like structures (LLSs) of HepG2 cells. These results can provide some data and reference in the understanding of the interaction between hemorheological properties and tumor vascular structures in solid tumors.


Assuntos
Reatores Biológicos , Técnicas de Cultura de Células/instrumentação , Separação Celular/instrumentação , Células Hep G2/fisiologia , Dispositivos Lab-On-A-Chip , Neovascularização Patológica/fisiopatologia , Desenho de Equipamento , Análise de Falha de Equipamento , Células Hep G2/patologia , Humanos , Neovascularização Patológica/patologia
15.
Biochem Biophys Res Commun ; 495(1): 1432-1439, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29158087

RESUMO

Functional abnormal airway epithelial cells, along with activated inflammatory cells, resulting in chronic airway inflammation, are considered as the characteristic of asthma. Fatty Acid Binding Protein 4 (FABP4) takes part in glucose and lipid homeostasis, and also have an important role in allergic airway inflammation. However, whether FABP4 influence barrier function of airway epithelial cells is unknown. In vivo, a HDM-induced murine model of asthma was obtained to assessed airway inflammation and protein expression of E-cadherin and Forkhead Box M1 (FoxM1). In vitro, 16-HBE was cultured and was treated with hrFABP4, siFABP4, FABPF4 inhibitor BMS, or FoxM1 inhibitor RCM-1. IL-4, IL-5, and IL-13 level was determined by ELISA. Transepithelial electrical resistance (TER), paracellular permeability and E-cadherin-special immunofluorescence were measured to value airway epithelial barrier function. Intracellular ROS production was determined by DCF-DA fluorescence. FABP4 inhibitor BMS alleviate airway inflammation and destruction of E-cad in allergic mouse. Treatment with HDM or hrFABP4 aggravated inflammatory response, damaged airway epithelial barrier, which could be inhibited by siFABP4 and BMS. Treatment with HDM or hrFABP4 also enhanced levels of FoxM1, and Inhibited FoxM1 suppressed HDM- and hrFABP4-induced inflammation and airway epithelial barrier dysfunction. In addition, H2O2 promoted FoxM1 expression, HDM and hrFABP4 induced-FoxM1 could be inhibited by NAC, leading to decreased inflammation and improved airway epithelial barrier. Upregulated ROS induced by FABP4 was of significance in activating FoxM1 leading to airway inflammation and epithelial barrier dysfunction.


Assuntos
Células Epiteliais Alveolares/imunologia , Asma/imunologia , Permeabilidade da Membrana Celular/imunologia , Proteínas de Ligação a Ácido Graxo/imunologia , Proteína Forkhead Box M1/imunologia , Espécies Reativas de Oxigênio/imunologia , Mucosa Respiratória/imunologia , Animais , Asma/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mucosa Respiratória/patologia
16.
Respir Res ; 19(1): 27, 2018 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-29422044

RESUMO

BACKGROUND: Endothelial cell (EC) regeneration is essential for inflammation resolution and vascular integrity recovery after inflammatory vascular injury. Cdc42 is a central regulator of cell survival and vessel formation in EC development. However, it is unknown that whether Cdc42 could be a regulating role of EC repair following the inflammatory injury in the lung. The study sought to test the hypothesis that Cdc42 is required for endothelial regeneration and vascular integrity recovery after LPS-induced inflammatory injury. METHODS AND RESULTS: The role of Cdc42 for the regulation of pulmonary vascular endothelial repair was tested in vitro and in vivo. In LPS-induced acute lung injury (ALI) mouse models, knockout of the Cdc42 gene in ECs increased inflammatory cell infiltration and pulmonary vascular leakage and inhibited vascular EC proliferation, which eventually resulted in more severe inflammatory lung injury. In addition, siRNA-mediated knockdown of Cdc42 protein on ECs disrupted cell proliferation and migration and tube formation, which are necessary processes for recovery after inflammatory vascular injury, resulting in inflammatory vascular injury recovery defects. CONCLUSION: We found that Cdc42 deficiency impairs EC function and regeneration, which are crucial in the post-inflammatory vascular injury repair process. These findings indicate that Cdc42 is a potential target for novel treatments designed to facilitate endothelial regeneration and vascular repair in inflammatory pulmonary vascular diseases, such as ALI/ARDS.


Assuntos
Endotélio Vascular/fisiologia , Regeneração/fisiologia , Lesões do Sistema Vascular/metabolismo , Proteína cdc42 de Ligação ao GTP/deficiência , Animais , Movimento Celular/fisiologia , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/patologia , Proteína cdc42 de Ligação ao GTP/genética
17.
Am J Physiol Lung Cell Mol Physiol ; 313(6): L1006-L1015, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28860147

RESUMO

Pulmonary fibrosis is characterized by lung fibroblast activation and ECM deposition and has a poor prognosis. Heat shock protein 90 (Hsp90) participates in organ fibrosis, and extracellular Hsp90α (eHsp90α) promotes fibroblast activation and migration. This study aimed to investigate whether a selective anti-Hsp90α monoclonal antibody, 1G6-D7, could attenuate lung fibrosis and whether 1G6-D7 presents a protective effect by inactivating the profibrotic pathway. Our results showed that eHsp90α was increased in mice with BLM-induced pulmonary fibrosis and that 1G6-D7 attenuated inflammation and collagen deposition in the lung. TGF-ß1 induced eHsp90α secretion, concomitantly promoting HFL-1 activation and ECM synthesis. 1G6-D7-mediated inhibition of eHsp90α significantly blocked these effects, meanwhile inhibiting downstream profibrotic pathways such as ERK, Akt, and P38. Human recombinant (hr)Hsp90α mimicked the effects of TGF-ß1, by activating profibrotic pathways and by upregulating LRP-1. Moreover, ERK inhibition effectively blocked the effect of (hr)Hsp90α. In conclusion, 1G6-D7 significantly protects against BLM-induced pulmonary fibrosis by ameliorating fibroblast activation and ECM production, which may be through blocking ERK signaling. Our results suggest a safer molecular therapy, 1G6-D7, in pulmonary fibrosis.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fibrose Pulmonar/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Bleomicina/efeitos adversos , Bleomicina/farmacologia , Linhagem Celular , Proteínas de Choque Térmico HSP90/imunologia , Humanos , Sistema de Sinalização das MAP Quinases/imunologia , Camundongos , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/patologia , Fator de Crescimento Transformador beta1/imunologia
18.
Cell Immunol ; 312: 15-24, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27884393

RESUMO

BACKGROUND: In our previous studies, we have indentified that 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) can alleviate toluene diisocyanate-induced airway epithelial barrier disruption and we also found that vascular endothelial growth factor (VEGF) derived from airway epithelials cells could disrupt epithelial barrier. OBJECTIVE: The study aimed to investigate whether 1,25(OH)2D3 can inhibit house dust mite (HDM) induced airway epithelial barrier dysfunction by regulating the VEGF pathway. METHOD: The 16HBE and BEAS-2B cells were cultured and treated according to the experiment requirement. Trans Epithelial Electric Resistance (TEER), permeability of epithelial layer, and distribution and expression of junction proteins were used to evaluate the cell layer barrier function, Western Blot was used to evaluate the expression of junction proteins and phosphorylated Akt in the cells, RT-PCR and ELISA were used to evaluate the VEGF gene expression and protein release in the cells. Recombinant VEGF165 was used to determine the role of the VEGF pathway in the epithelial barrier function. RESULTS: HDM resulted in a decline in TEER and increase of cell permeability, following abnormal distribution and expression of junction proteins (E-Cadherin and zona occludens (ZO)-1), accompanied by a significant upregulation of VEGF and phosphorylated Akt, which were all partly recovered by treatment with either 1,25(OH)2D3 or PI3K inhibitor LY294002. VEGF165-induced barrier dysfunction was accompanied by disruption of the epithelial E-cadherin and ß-catenin, pretreatment of 1,25(OH)2D3 and LY294002 markedly attenuated VEGF-induced airway barrier disruption in 16HBE cells. CONCLUSION: 1,25(OH)2D3 can alleviate HDM-induced airway epithelial barrier dysfunction by inhibiting PI3K pathway-dependent VEGF release.


Assuntos
Antialérgicos/farmacologia , Calcitriol/farmacologia , Hipersensibilidade/tratamento farmacológico , Mucosa Respiratória/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Antígenos de Dermatophagoides/imunologia , Caderinas/metabolismo , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Impedância Elétrica , Humanos , Hipersensibilidade/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Pyroglyphidae , Mucosa Respiratória/patologia , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , beta Catenina/metabolismo
19.
Cell Immunol ; 315: 56-63, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28400057

RESUMO

Recent studies have indicated that Thymic stromal lymphopoietin (TSLP) plays an important role in the prevention and treatment of asthma. However the role of TSLP in dysfunction of airway epithelial adherens junctions E-cadherin in house dust mite (HDM)-induced asthma has not been addressed. We hypothesized that TSLP contributed to HDM-induced E-cadherin dysfunction in asthmatic BALB/c mice and 16HBE cells. In vivo, a HDM-induced asthma mouse model was set up for 8weeks. Mice inhaled an anti-TSLP monoclonal antibody (mAb) before HDM. The mice treated with the anti-TSLP mAb ameliorated airway inflammation, the decreasing and aberrant distribution of E-cadherin and ß-catenin as well as phosphorylation(p)-AKT induced by HDM. In vitro, HDM increased the expression of TSLP and E-cadherin dysfunction by PI3K/Akt signaling pathway. The exposure of 16HBE to TSLP resulted in redistribution of E-cadherin. These results indicate that TSLP may be an important contributor in E-cadherin dysfunction of HDM-induced asthma. TSLP signaling blocking shows a protective effect in mice and that the PI3K/Akt pathway may play a role in this process.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Asma/imunologia , Caderinas/metabolismo , Citocinas/fisiologia , Pyroglyphidae/imunologia , Administração por Inalação , Animais , Anticorpos Monoclonais/administração & dosagem , Asma/terapia , Brônquios/citologia , Hiper-Reatividade Brônquica/etiologia , Hiper-Reatividade Brônquica/imunologia , Hiper-Reatividade Brônquica/prevenção & controle , Linhagem Celular , Cromonas/farmacologia , Citocinas/antagonistas & inibidores , Citocinas/biossíntese , Citocinas/imunologia , Modelos Animais de Doenças , Células Epiteliais , Humanos , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Morfolinas/farmacologia , Proteína Oncogênica v-akt/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Fosforilação , Processamento de Proteína Pós-Traducional , Distribuição Aleatória , Transdução de Sinais/imunologia , Organismos Livres de Patógenos Específicos , beta Catenina/análise , Linfopoietina do Estroma do Timo
20.
Microvasc Res ; 109: 45-53, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27769693

RESUMO

BACKGROUND: After stimulation due to injury, cell division cycle protein 42 (Cdc42) restores and enhances barrier functions by strengthening intercellular adherens junctions; however, its influence on cell proliferation after injury remains unknown. OBJECTIVE: In this study, we sought to investigate the effect of stimulation using small doses of lipopolysaccharide (LPS) on the proliferation of pulmonary microvascular endothelial cells (PMVECs). METHODS: We stimulated PMVECs with different doses of LPS and evaluated the effects on cell proliferation. We also constructed a primary gene-knockout cell line lacking Cdc42 to verify the role of Cdc42 in regulating the proliferation of PMVECs that were stimulated using LPS and to explore related signaling pathways. RESULTS: Stimulating PMVECs with small doses of LPS increased proliferation. Cdc42 is involved in regulating this process, which was mediated by the extracellular regulated protein kinase (ERK) pathway. CONCLUSIONS: Cdc42 plays a role in regulating the proliferation of PMVECs stimulated with small doses of LPS, and this regulation involves the ERK pathway.


Assuntos
Células Endoteliais/citologia , Pulmão/citologia , Sistema de Sinalização das MAP Quinases , Microcirculação , Proteína cdc42 de Ligação ao GTP/metabolismo , Animais , Proliferação de Células , Endotélio Vascular/citologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Lipopolissacarídeos , Pulmão/irrigação sanguínea , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microvasos/citologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA