Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Redox Rep ; 27(1): 139-149, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35801580

RESUMO

OBJECTIVES: Sevoflurane is identified as an effective candidate drug for acute lung injury (ALI) treatment, but its curing effects and detailed mechanisms have not been fully disclosed. The present study was designed to resolve this academic issue. METHODS: The ALI mice models were established, and Hematoxylin-eosin staining assay was performed to examine tissue morphologies. Cell viability was determined by CCK-8 assay, and Annexin V-FITC/PI double staining assay was used to examine cell apoptosis. The expression levels of proteins were determined by performing Western Blot analysis and immunofluorescence staining assay. ROS levels were examined by using DCFH-DA staining assay. RESULTS: In this study, we investigated this issue and the ALI models were respectively established by treating the BALB/c mice and the murine macrophage cell line RAW264.7 with different concentrations of lipopolysaccharide (LPS) in vivo and in vitro, which were subsequently subjected to sevoflurane co-treatment. The results showed that sevoflurane reduced LPS-induced ALI in mice and suppressed LPS-triggered oxidative stress and apoptotic cell death in the RAW264.7 cells. Interestingly, we evidenced that the elimination of reactive oxygen species (ROS) by N-acetyl-L-cysteine (NAC) reversed LPS-induced cell apoptosis in RAW264.7 cells. Then, we verified that sevoflurane suppressed oxidative damages to restrain LPS-induced apoptotic cell death in the RAW264.7 cells through activating the anti-oxidant Keap1/Nrf2 pathway. Mechanistically, sevoflurane down-regulated Keap1 and upregulated Nrf2 in nucleus to activate the downstream anti-oxidant signaling cascades, which further ameliorated LPS-induced cell apoptosis and lung injury by eliminating oxidative damages. DISCUSSION: Taken together, our study illustrated that the sevoflurane attenuates LPS-induced ALI by inhibiting oxidative stress-mediated apoptotic cell death and inflammation, and the Keap1/Nrf2 pathway played an important role in this process.


Assuntos
Lesão Pulmonar Aguda , Estresse Oxidativo , Fibrose Pulmonar , Sevoflurano , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/metabolismo , Animais , Antioxidantes/metabolismo , Apoptose , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Lipopolissacarídeos , Pulmão/metabolismo , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Células RAW 264.7 , Espécies Reativas de Oxigênio/metabolismo , Sevoflurano/uso terapêutico
2.
Int Immunopharmacol ; 109: 108800, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35550264

RESUMO

Pyroptosis is a type of programmed cell death, and pyroptosis-associated inflammatory response is closely associated with the pathogenesis of acute lung injury (ALI). Sevoflurane, a common clinical anesthetic, has been reported as therapeutic drug for ALI. However, the detailed mechanisms by which sevoflurane ameliorates ALI have not been fully delineated. In this study, we found that sevoflurane phosphorylated and activated the GSK-3ß to suppress LPS-induced pyroptotic cell death, inflammation and ALI. Specifically, in the LPS-induced ALI mice models, sevoflurane attenuated lung damages and fibrosis, and restrained the production of the pro-inflammatory cytokines. Also, LPS increased the expression levels of pyroptosis-related proteins to promote pyroptotic cell death in ALI mice lung tissues, and LPS-induced pyroptotic cell death was reduced by sevoflurane co-treatment. Moreover, the potential underlying mechanisms were uncovered, and we illustrated that sevoflurane promoted GSK-3ß activation in LPS-treated ALI mice lung tissues, and re-activation of GSK-3ß by the PI3K/Akt pathway inhibitor LY294002 suppressed LPS-induced pyroptotic cell death in vivo. Consistently, in the in vitro macrophages, our data hinted that LPS-induced pyroptotic cell death were also reversed by sevoflurane. Collectively, the above results suggest that sevoflurane re-activated GSK-3ß to suppress LPS-induced pyroptotic cell death, inflammation and ALI.


Assuntos
Lesão Pulmonar Aguda , Lipopolissacarídeos , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/metabolismo , Animais , Glicogênio Sintase Quinase 3 beta/metabolismo , Inflamassomos/metabolismo , Inflamação/tratamento farmacológico , Lipopolissacarídeos/farmacologia , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Piroptose , Sevoflurano/uso terapêutico
3.
Int Immunopharmacol ; 108: 108869, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35605434

RESUMO

BACKGROUND: Sevoflurane anesthesia is deemed as potential therapeutic drug for lipopolysaccharide (LPS)-induced acute lung injury (ALI), but the molecular mechanisms have not been fully delineated. AIM: The present study explored the specific molecular mechanism of sevoflurane regulating autophagy to reduce LPS induced ALI. METHODS: Male C57BL/6J mice and mouse pulmonary microvascular endothelial cells (MPVECs) were treated with LPS to construct ALI models, and the levels of inflammation, apoptosis and autophagy were detected after treatment with sevoflurane. Meanwhile, cells were treated with autophagy inhibitor or AMP-activated protein kinase (AMPK)/unc-51 like autophagy activating kinase 1 (ULK1) pathway inhibitor in vitro to detect their effects on cell survival. RESULTS: Sevoflurane reduced inflammation, recovered cell division so as to suppress cell apoptosis and maintain cell survival, and activated autophagic flux in LPS-induced ALI models in vivo and in vitro. Of note, the suppressing effects of sevoflurane on LPS-induced cell death were abrogated by inhibiting autophagy. Moreover, we evidenced that sevoflurane promoted activation of the AMPK/ULK1 pathway in LPS-induced ALI models. Blockage of this pathway abrogated the promoting effects of sevoflurane on cell autophagy and cell viability in LPS-treated cells. CONCLUSION: Collectively, sevoflurane suppresses apoptosis and inflammation via activating protective autophagy, thereby ameliorating LPS-induced ALI, and the AMPK/ULK1/ PIKFYVE pathway is responsible for the process.


Assuntos
Lesão Pulmonar Aguda , Anestesia , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/metabolismo , Animais , Autofagia , Células Endoteliais/metabolismo , Inflamação , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/metabolismo , Sevoflurano/farmacologia , Sevoflurano/uso terapêutico , Transdução de Sinais
4.
BMC Pulm Med ; 22(1): 159, 2022 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-35473680

RESUMO

BACKGROUND: Sevoflurane is considered as a lung-protective factor in acute lung injury (ALI), but the underlying molecular mechanism remains largely unknown. The present study identified for the first time that sevoflurane ameliorated lipopolysaccharide (LPS)-induced ALI through regulating a novel long non-coding RNA LINC00839, and uncovered its regulatory mechanism. METHODS: LPS-induced ALI models were established in mice or mouse pulmonary microvascular endothelial cells (MPVECs), and they were administered with sevoflurane. Real-Time quantitative PCR, western blot and bioinformatics analysis were performed to screen the aberrantly expressed long non-coding RNA and the downstream molecules in sevoflurane-treated ALI models, and their roles in the protection effect of sevoflurane were verified by functional recovery experiments. RESULTS: Sevoflurane relieved LPS-induced lung injury, cell pyroptosis and inflammation in vitro and in vivo. LINC00839 was significantly suppressed by sevoflurane, and overexpression of LINC00839 abrogated the protective effects of sevoflurane on LPS-treated MPVECs. Mechanismly, LINC00839 positively regulated NOD-like receptor protein 3 (NLRP3) via sequestering miR-223. MiR-223 inhibitor reversed the inhibitory effects of LINC00839 knockdown on NLRP3-mediated pyroptosis in LPS-treated MPVECs. Furthermore, both miR-223 ablation and NLRP3 overexpression abrogated the protective effects of sevoflurane on LPS-treated MPVECs. CONCLUSION: In general, our work illustrates that sevoflurane regulates the LINC00839/miR-223/NLRP3 axis to ameliorate LPS-induced ALI, which might provide a novel promising candidate for the prevention of ALI.


Assuntos
Lesão Pulmonar Aguda , Anestesia , MicroRNAs , RNA Longo não Codificante , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/genética , Animais , Células Endoteliais/metabolismo , Humanos , Lipopolissacarídeos/farmacologia , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteínas NLR , RNA Longo não Codificante/genética , Sevoflurano/efeitos adversos
5.
Cell Cycle ; 19(17): 2235-2246, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32794418

RESUMO

Circular RNA (circRNA) is currently considered to be a key regulatory molecule in cancer biology. In the present study, we aimed to explore the functional and clinical roles of circ-SLC7A6 (a circRNA derived from SLC7A6 gene) in non-small cell lung cancer (NSCLC). Circ-SLC7A6 was significantly downregulated in NSCLC tissues in comparison to para-carcinoma tissues. Low circ-SLC7A6 was closely associated with larger tumor size, lymph node metastasis, advanced clinical stage and adverse outcome. Exogenous expression of circ-SLC7A6 evidently inhibited the proliferation and invasion of NSCLC cells. Further investigations revealed that miR-21 was the direct functional target of circ-SLC7A6, in which circ-SLC7A6 abundantly sponged miR-21 and elevated a cohort of tumor suppressors, thus inhibiting NSCLC progression. Interestingly, QKI, elevated by circ-SLC7A6, could directly bind to the introns flanking circ-SLC7A6 to facilitate circ-SLC7A6 production. Importantly, in vivo xenograft tumor experiments showed that reintroduction of circ-SLC7A6 retarded tumor growth as well as decreased lung metastatic nodules. Overall, our study demonstrates that circ-SLC7A6 is a novel tumor suppressor in NSCLC, targeting circ-SLC7A6/miR-21 axis may be a promising treatment for NSCLC patients.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Genes Supressores de Tumor , Neoplasias Pulmonares/genética , MicroRNAs/metabolismo , RNA Circular/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/patologia , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Invasividade Neoplásica , Metástase Neoplásica , RNA Circular/genética , Proteínas de Ligação a RNA/metabolismo
6.
Mater Sci Eng C Mater Biol Appl ; 112: 110786, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32409023

RESUMO

The aim of this study was to prepare a promising drug carrier for treatment of lung cancer. The self-assembly nanoparticles of SDP-GEM/PEI-PEG-anti-EGFR with chemotherapeutic drug of gemcitabine (GEM), Magnetic resonance imaging (MRI) guided- imaging and targeting of anti- Epidermal Growth Factor Receptor (anti-EGFR) were designed. The imaging capacity, targeting feasibility and anti-tumor function were evaluated respectively. SDP-GEM/PEI-PEG-anti-EGFR exhibited contrast enhancement under T2 Weight Image (T2WI) and a liner relationship was found between the concentration and relaxation rate of R2 and R2* in vitro. With the targeting of anti-EGFR, the endocytosis of nanoparticles increased significantly, which effectively killed lung cancer cells in vitro, and importantly it can be accurately delivered to tumor site within 3 h in vivo. Prolonged lifetime and smaller tumor volume demonstrated that SDP-GEM/PEI-PEG-anti-EGFR efficiently inhibited tumor growth in vivo. Therefore, SDP-GEM/PEI-PEG-anti-EGFR was an effective and safe drug carrier, which had a great potential application in MRI-guided lung cancer therapy.


Assuntos
Materiais Biocompatíveis/uso terapêutico , Desoxicitidina/análogos & derivados , Portadores de Fármacos/química , Neoplasias Pulmonares/tratamento farmacológico , Nanopartículas/química , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Materiais Biocompatíveis/química , Materiais Biocompatíveis/metabolismo , Materiais Biocompatíveis/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Desoxicitidina/química , Desoxicitidina/metabolismo , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Endocitose , Receptores ErbB/imunologia , Feminino , Hemólise/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Nanopartículas/metabolismo , Polietilenoglicóis/química , Polietilenoimina/análogos & derivados , Polietilenoimina/química , Pró-Fármacos/química , Pró-Fármacos/metabolismo , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Distribuição Tecidual , Transplante Heterólogo , Gencitabina
7.
Biomed Pharmacother ; 124: 109828, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31986409

RESUMO

Recently, increasing studies have confirmed some circRNAs were involved in the genesis of chemotherapy resistance in almost all kinds of malignant tumors, including lung adenocarcinoma (LAD). Nevertheless, the function and mechanism of circPVT1 in regulating chemotherapy resistance of LAD has not been elucidated so far. The current study found circPVT1 was highly expressed in LAD, which expression was positively related to N stage and chemotherapy insensitivity (cisplatin and pemetrexed) of LAD patients, and it was an independent prognostic biomarker for LAD patients. The circPVT1 expression was up-regulated in LAD tissues and cell line (A549/DR) resistant to cisplatin and pemetrexed. CircPVT1 knockdown sensitized A549/DR cells to cisplatin and pemetrexed. RNA pull-down assay et al. confirmed circPVT1 acted as a ceRNA for miR-145-5p in A549/DR cells. In addition, miR-145-5p was lowly expressed in cisplatin and pemetrexed resistant LAD tissues and cell line, and its over-expression also sensitized A549/DR cells to cisplatin and pemetrexed. The luciferase reporter assay et al. proved ABCC1 was a target gene of miR-145-5p in A549/DR cells. Moreover, miR-145-5p enhancement partly restored the effecting of circPVT1 knockdown on chemotherapy resistance in A549/DR cells, miR-145-5p/ABCC1 pathway mediated chemotherapy resistance induced by circPVT1 knockdown in LAD cells. In conclusion, the high-expression of circPVT1 is related with the cisplatin and pemetrexed insensitivity of LAD patients, circPVT1 contributes to cisplatin and pemetrexed chemotherapy resistance through miR-145-5p/ABCC1 axis.


Assuntos
Adenocarcinoma de Pulmão/tratamento farmacológico , Antineoplásicos/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , RNA Longo não Codificante/genética , Células A549 , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/patologia , Biomarcadores Tumorais/genética , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Pemetrexede/farmacologia , Regulação para Cima
8.
Clin Respir J ; 14(2): 173-178, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31794151

RESUMO

Cardiac epithelioid hemangioendothelioma (EHE) is a very rare tumour of endothelial origin with the lung and liver as the most easily metastatic organs. We describe herein a patient with hemoptysis, severe anaemia, and diffuse pulmonary nodules with halo signs that represented metastasis of cardiac EHE; these radiologic manifestations are relatively uncommon. During the initial workup for the patient's pulmonary nodules, echocardiography missed the cardiac mass. However, positron emission tomography-computed tomography revealed increased fluorodeoxyglucose intake in the right atrial wall, and cardiac magnetic resonance imaging (MRI) revealed an irregular nodule with normal T1-weighted signal intensity and hyperintense T2-weighted signal intensity. Enhanced abdominal computed tomography (CT) revealed micronodular liver metastases. Video-assisted thoracic surgery was performed to make a definitive diagnosis. Immunohistochemistry staining proved the diagnosis of EHE with positive results for cluster of differentiation (CD) 34, CD31, erythroblast transformation-specific-related gene and Ki-67. The patient started chemotherapy with docetaxel (75 mg/m2 ) and gemcitabine (900 mg/m2 ), but this failed to control his disease and he died from an opportunistic infection related to his immunocompromised status 5 months later. For the work out process of bilateral diffuse pulmonary nodules suspicious for cardiac origin, especially with atrial deviation, echocardiography alone is not sufficient to exclude atrial origin. Cardiac CT or MRI might be a better choice.


Assuntos
Neoplasias Cardíacas/diagnóstico , Hemangioendotelioma Epitelioide/secundário , Neoplasias Pulmonares/secundário , Diagnóstico Diferencial , Átrios do Coração , Hemangioendotelioma Epitelioide/diagnóstico , Humanos , Neoplasias Pulmonares/diagnóstico , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Cirurgia Torácica Vídeoassistida
9.
Oncol Rep ; 42(6): 2402-2415, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31638248

RESUMO

The competing endogenous RNA (ceRNA) network is crucial for the development and progression of tumors, including non­small cell lung cancer (NSCLC). However, what type of ceRNA network regulates NSCLC has not been clarified. The present study aimed to elucidate the long non­coding RNA (lncRNA)/microRNA (miRNA)/mRNA ceRNA network in NSCLC, particularly for the significance of lncRNAs in NSCLC. NSCLC­specific differentially expressed lncRNAs, miRNAs and mRNAs in the Cancer Genome Atlas (TCGA) were analyzed and their relationship was analyzed by a ceRNA network. Their potential functions of differentially expressed mRNAs were analyzed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Furthermore, the expression levels of four selected lncRNAs in TCGA were determined and their associated survival of patients was examined. In addition, the expression profiles of these four lncRNAs in 48 NSCLC specimens and cell lines, their cellular distribution and associated clinical parameters were examined. We successfully constructed a ceRNA network, including 113 lncRNAs, 12 miRNAs and 36 mRNAs differentially expressed between NSCLC and non­tumor tissues. LINC00525, MED4­AS1, STEAP2­AS1 and SYNPR­AS1 lncRNAs were selected and validated for their association with the survival of NSCLC patients. The expression of these lncRNAs was upregulated in 48 NSCLC tissues and was varying in NSCLC cells. While LINC00525 was mainly expressed in the cytoplasm, MED4­AS1 was in both the nucleus and cytoplasm of A549 cells. In addition, the expression of LINC00525 was significantly associated with smoking history (P<0.05); MED4­AS1 was significantly associated with women, poor differentiation and lymph node metastasis (P<0.05); STEAP2­AS1 was significantly associated with women (P<0.01); and SYNPR­AS1 was significantly associated with women and adenocarcinoma (P<0.05). These lncRNAs may be valuable biomarkers for prognosis of NSCLC and the ceRNA network may provide new insights in the pathogenesis of NSCLC.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , MicroRNAs/genética , RNA Longo não Codificante/genética , RNA Mensageiro/genética , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/secundário , Idoso , Idoso de 80 Anos ou mais , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/secundário , Feminino , Seguimentos , Ontologia Genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Prognóstico , Taxa de Sobrevida , Transcriptoma
10.
J Cardiothorac Surg ; 14(1): 60, 2019 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-30871612

RESUMO

BACKGROUND: Atrial fibrillation (AF) is a common complication after radical surgery of esophageal cancer. The aim of this study was to explore AF risk factors after radical surgery of esophageal carcinoma. METHOD: The data of 335 patients with esophageal cancer who were admitted in our hospital from January 2014 to August 2016 for the first time were retrospectively analyzed. We retrieved the papers in some data banks using the search terms including English and Chinese search terms, and obtained 13 factors which were mentioned in more than 6 papers. The 13 factors including age, gender, history of smoking, history of hypertension, history of peripheral vascular disease, history of cardiac stents or angina pectoris, preoperative pulmonary infection, preoperative brain natriuretic peptide (BNP) level, preoperative left ventricular diastolic dysfunction, operative method, lesion location, intraoperative blood transfusion, adhesion between lymph nodes and pericardium, underwent univariate and multivariate analyses. RESULTS: Of the 335 patients with esophageal cancer, 48 had AF within one week after operation. Univariate analysis indicated that the age (OR: 4.89; CI: 2.53-9.47, P: 0.000), gender (OR: 2.26; CI: 1.17-4.37, P: 0.013), history of peripheral vascular disease (OR: 2.29; CI: 1.06-4.92, P: 0.030), history of cardiac stents or angina pectoris (OR: 27.30; CI: 12.44-59.91, P: 0.000), preoperative BNP level (OR: 27.13; CI: 10.97-67.06, P: 0.000), preoperative left ventricular diastolic dysfunction (OR: 2.22; CI: 1.19-4.14, P: 0.012), operative method (OR: 2.09; CI: 1.002-4.380, P: 0.046), intraoperative blood transfusion (OR: 20.24; CI: 8.39-48.82, P: 0.000), and adhesion between lymph nodes and pericardium were risk factors (OR: 2.05; CI: 1.08-3.87, P: 0.024). Furthermore, multivariate analysis displayed that advanced age (OR: 5.044; CI: 1.748-14.554, P: 0.003), male (OR: 6.161; CI: 2.143-17.715, P: 0.001), history of cardiac stents or angina pectoris (OR: 48.813; CI: 13.674-174.246, P: 0.000), preoperative BNP > 100 (OR: 41.515; CI: 9.380-183.732, P: 0.000), open surgery (OR: 3.357; CI: 1.026-10.983, P: 0.045), intraoperative blood transfusion (OR: 58.404; CI: 10.777-316.509, P: 0.000), and adhesion between lymph nodes and pericardium (OR: 3.954; CI: 1.364-11.459, P: 0.011) were risk factors which could increase the incidence of postoperative AF. CONCLUSION: We should pay attention to the above risk factors in order to reduce the incidence of postoperative AF.


Assuntos
Fibrilação Atrial/etiologia , Procedimentos Cirúrgicos do Sistema Digestório/efeitos adversos , Neoplasias Esofágicas/cirurgia , Complicações Pós-Operatórias/etiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Fatores de Risco
11.
J Thorac Dis ; 11(12): 5535-5546, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32030273

RESUMO

BACKGROUND: Non-small cell lung cancer (NSCLC) accounts for about 80-85% of lung cancers. Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) such as gefitinib are considered the best choice for first-line treatment for the patients with NSCLC harboring EGFR-activating alterations. Nonetheless, 10-30% of patients may not obtain an objective response and may also experience rapid progression. The aim of our research, based on the integrative bioinformatics review, was to identify the possible miRNAs involved in gefitinib resistance. METHOD: A gefitinib-resistant network composed of 15 miRNAs and 34 targets were constructed by using the bioinformatics analyses of three microarray datasets. Of these miRNAs, effects of miR-342-3p on gefitinib resistance were investigated on a gefitinib-resistant cell model (A549/GR and PC/GR cells). RESULTS: We reported that over-expression of miR-342-3p could significantly increase the resistance to gefitinib of A549/GR and PC9/GR cells and vice versa. Then, we recognized CPA4 as a target of hsa-miR-342-3p by a luciferase reporter assay. The increase in hsa-miR-342-3p levels led to a significant reduction in CPA4 protein expression. However, the opposite results were observed upon miR-342-3p knockdown. Finally, we found that enforced CPA4 expression partially reversed miR-342-3p effects in A549/GR cells. CONCLUSIONS: Collectively, these findings suggest that the upregulation of miR-342-3p contributes to gefitinib resistance by targeting CPA4, which may serve as a potential treatment option to overcome gefitinib resistance in patients with NSCLC.

12.
Gene ; 633: 61-65, 2017 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-28843520

RESUMO

The myocardial infarction associated transcript (MIAT), a long non-coding RNA (lncRNA), was originally identified as a candidate gene for myocardial infarction, and was recently shown to participate in the progression of cancer and the process of metastasis. However, the biological role of MIAT and the underlying mechanisms that mediate its role in non-small-cell lung cancer (NSCLC) remain unclear. Here, we have shown that the expression of MIAT in NSCLC tissues was upregulated. Knockdown of MIAT substantially inhibited the invasive ability of NSCLC cells. Moreover, the knockdown of MIAT significantly downregulated the expression of the zinc finger E-box binding homeobox 1 (ZEB1), that was upregulated in NSCLC and that promoted cell invasion. Rather than by direct interactions, we found that MIAT indirectly regulated ZEB1 expression through sponging and suppressing microRNA (miR)-150, which represses ZEB1 and interacts with MIAT in a sequence-specific manner. Thus, MIAT may inhibit ZEB1 expression and promote cell invasion of NSCLC cells via the miR-150/ZEB1 pathway. Taken together, our findings suggested that MIAT plays an oncogenic role in NSCLC through the ZEB1 signaling pathway by sponging miR-150, and MIAT may therefore serve as a valuable prognostic biomarker and therapeutic target for NSCLC.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/patologia , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo , Biomarcadores Tumorais/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Proliferação de Células , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Neoplasias Pulmonares/genética , Invasividade Neoplásica , Metástase Neoplásica , RNA Longo não Codificante/genética , Regulação para Cima , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética
13.
Biomed Pharmacother ; 90: 650-658, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28415044

RESUMO

The long non-coding RNAs (lncRNAs) have been recently shown to participate in the progression of a variety of cancers. However, the biological role of lncRNAs and the underlying mechanisms in Lung squamous cell carcinoma (SCC) or lung adenocarcinoma (AD) remain unclear. Herein, we investigated expression of 5 lncRNAS (SNHG1, NCBP2-AS2, LINC01206, SOX2-OT and LINC01419) in SCC and AD tissues. SNHG1 was one of over-expressed lncRNAs in SCC tissues. Knockdown of SNHG1 significantly inhibited the proliferation, metastasis, invasive ability and induced apoptosis of SCC cells. Moreover, SNHG1 affected the expression of zinc finger E-box binding homeobox 1(ZEB1), which has also been observed to be up-regulated in SCC and to promote cell metastasis and invasion. Rather than direct interaction, SNHG1 regulated ZEB1expression by suppressing the activity of p63 TA isoform (TAp63), which is a repressor of ZEB1 and physically associates with SNHG1. Furthermore, SNHG1 promoted ZEB1 expression and promoted cell proliferation, metastasis, invasive but inhibited apoptosis of SCC cells via the TAp63/ZEB1 pathway. Taken together, our findings suggested that SNHG1 might play an oncogenic role in SCC through ZEB1 signaling pathway by inhibiting TAp63 and might serve as a valuable prognostic biomarker and therapeutic target for SCC patients.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma de Células Escamosas/genética , Invasividade Neoplásica/genética , Metástase Neoplásica/genética , RNA Longo não Codificante/genética , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Idoso , Apoptose/genética , Biomarcadores Tumorais/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Escamosas/patologia , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Invasividade Neoplásica/patologia , Metástase Neoplásica/patologia , Transdução de Sinais/genética , Regulação para Cima/genética
14.
Biomed Pharmacother ; 69: 361-6, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25661383

RESUMO

Lung cancer is the leading cause of cancer-related death worldwide, mainly due to its highly metastatic properties. Previously, we reported an inverse correlation between Rho-kinase inhibitor and the progression of the lung cancer cells. The purpose of this study was to investigate the effects of RhoA on the proliferation, adhesion, invasion, and migration of SPCA1 lung carcinoma cells and to explore the underlying molecular mechanisms. RNA interference was used to downregulate RhoA expression in these cells. Through G418 screening, we generated SPCA1 lung cancer cell lines with stable RhoA silencing. We then observed the cell behaviour, and used matrix metalloproteinase (MMP) activity and western blot assays to evaluate the underlying molecular mechanisms. The proliferation, adhesion, migration, and invasion of SPCA1 lung cancer cells were decreased after knockdown of RhoA. At the molecular level, the total amounts of active MMP2 and MMP9 were decreased by about 17.21% (P<0.05) and 45.32% (P<0.01), respectively. Myosin phosphatase targeting subunit 1 phosphorylation (P-MYPT1) was reduced by 36.16% (P<0.05). Taken together, our findings show that the knockdown of RhoA prevents proliferation and metastasis in SPCA1 lung cancer cells. Changes in MMP2, MMP9, and P-MYPT1 levels and activity might be some of the molecular mechanisms underlying these effects.


Assuntos
Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/patologia , Proteína rhoA de Ligação ao GTP/metabolismo , Sequência de Bases , Western Blotting , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação para Baixo , Inativação Gênica , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Dados de Sequência Molecular , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Invasividade Neoplásica , Metástase Neoplásica , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...