Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Altern Ther Health Med ; 28(7): 34-39, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35951060

RESUMO

Objective: Aerosol inhalation is commonly used in the treatment of chronic obstructive pulmonary emphysema (COPE). This study aimed to evaluate the effectiveness of aerosol inhalation combined with a vibration expectoration machine on COPE. Methods: From June 2019 to June 2020, 110 patients receiving COPE treatment in Linyi Central Hospital in China were included in this randomized controlled trial. All patients were randomly assigned into one of two groups using the random number table. A total of 55 patients were given aerosol inhalation combined with the use of a vibration expectoration machine in the study group, and 55 patients were given aerosol inhalation alone in the control group. The general data, clinical efficacy arterial blood gas index, pulmonary function index and serum levels of insulin-like growth factor 1 (IGF-1), alpha 1 antitrypsin (α1-AT) and platelet-derived growth factor-B (PDGF-B) were compared. Results: There was no difference in baseline characteristics between the 2 groups (P > .05). After treatment, the clinical efficacy in the study group was significantly higher than in the control group (96.36% vs 81.82%, respectively; P = .023), daily sputum production in the study group was significantly higher compared with the control group (80.92 ± 10.29 vs 58.63 ± 9.02 ml, respectively; P < .001) and hospitalization time was significantly reduced in the study group (11.87 ± 1.76 vs 17.62 ± 1.92 days, respectively; P < .001). In addition, the respiratory rate was significantly lower in the study group (17.43 ± 1.61 vs 22.08 ± 3.25, respectively; P < .001). Partial pressure of oxygen (P[O2]) was significantly higher (76.29 ± 7.34 vs 66.81 ± 7.93 mmHg, respectively; P < .001) and partial pressure of carbon dioxide (P[CO2]) was significantly lower (34.82 ± 6.02 vs 39.83 ± 6.01 mmHg respectively; P < .001) in the study group compared with the control group. In addition, forced expiratory volume in the first second (FEV1) (1.79 ± 0.36 vs 1.66 ± 0.28 L, respectively), forced vital capacity (FVC) (2.58 ± 0.28 vs 2.42 ± 0.11 L, respectively), forced expiratory volume in the first second as a percentage of the predicted value (FEV1%pred) (65.32 ± 4.03 vs 59.83 ± 4.76 L, respectively) and maximal mid-expiratory flow (MMEF) (1.51 ± 0.27% vs 1.36 ± 0.12%, respectively) were all significantly increased after treatment in the study group compared with the control group (all P < .001). The IGF-1 (104.92 ± 11.27 vs 137.83 ± 11.02 ng/mL, respectively) and PDGF-B (124.39 ± 14.29 vs 249.93 ± 33.49 ng/L, respectively) were significantly reduced in the study group after treatment (all P < .001). The α1-AT (2.82 ± 0.38 vs 2.17 ± 0.22 g/L, respectively) were significantly increased after treatment in the study group compared with the control group. Conclusion: Aerosol inhalation combined with the use of a vibration expectoration machine is worthy of clinical application, and can effectively improve outcomes in patients with COPE.


Assuntos
Doença Pulmonar Obstrutiva Crônica , Enfisema Pulmonar , Dióxido de Carbono/uso terapêutico , Volume Expiratório Forçado , Humanos , Fator de Crescimento Insulin-Like I/uso terapêutico , Oxigênio/uso terapêutico , Proteínas Proto-Oncogênicas c-sis/uso terapêutico , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Enfisema Pulmonar/tratamento farmacológico , Aerossóis e Gotículas Respiratórios , Escarro , Vibração/uso terapêutico , alfa 1-Antitripsina/uso terapêutico
2.
J Surg Res ; 263: 140-150, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33652176

RESUMO

BACKGROUND: Emerging evidence show that long noncoding RNAs (lncRNAs) are crucial regulators in pathophysiology of acute lung injury (ALI). Small nucleolar RNA host gene 14 (SNHG14) is a novel oncogenic lncRNA, and has been associated with inflammation-related cell injuries. Thus, we wondered the role and mechanism of SNHG14 in lipopolysaccharides (LPS)-induced ALI cell model. METHODS: Expression of SNHG14, miRNA (miR)-124-3p, and transforming growth factor ß type 2 receptor (TGFBR2) was detected by RT-qPCR and western blotting. Cell apoptosis was determined by methyl thiazolyl tetrazolium assay, flow cytometry, western blotting, and lactate dehydrogenase activity kit. Inflammation was measured by enzyme-linked immunosorbent assay. The interaction among SNHG14, miR-124-3p, and TGFBR2 was validated by dual-luciferase reporter assay and RNA immunoprecipitation. RESULTS: LPS administration attenuated human lung epithelial cell viability and B-cell lymphoma-2 expression, but augmented apoptosis rate, cleaved-caspase-3 expression, lactate dehydrogenase activity, and secretions of tumor necrosis factor-α, interleukin-1ß, and IL-6 in A549 cells. Thus, LPS induced A549 cells apoptosis and inflammation, wherein SNHG14 was upregulated and miR-124-3p was downregulated. However, silencing SNHG14 could suppress LPS-induced apoptosis and inflammation depending on upregulating miR-124-3p via target binding. Similarly, overexpressing miR-124-3p attenuated LPS-induced A549 cells injury through inhibiting its downstream target TGFBR2. Furthermore, SNHG14 knockdown could also affect TGFBR2 expression via miR-124-3p. CONCLUSIONS: SNHG14 knockdown prevents A549 cells from LPS-induced apoptosis and inflammation through regulating miR-124-3p and TGFBR2, suggesting a novel SNHG14/miR-124-3p/TGFBR2 circuit in alveolar epithelial cells on the set of ALI.


Assuntos
Lesão Pulmonar Aguda/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II/genética , Células A549 , Lesão Pulmonar Aguda/imunologia , Apoptose/genética , Apoptose/imunologia , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Regulação para Baixo/imunologia , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Lipopolissacarídeos/imunologia , MicroRNAs/agonistas , MicroRNAs/genética , RNA Longo não Codificante/genética , Regulação para Cima/imunologia
3.
Mol Med Rep ; 23(2)2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33300072

RESUMO

Non­small­cell lung cancer (NSCLC) accounts for 80% of lung cancer cases, and is the leading cause of cancer­associated mortality worldwide. The present study aimed to investigate the roles of microRNA (miR)­654­3p in NSCLC. The expression levels of miR­654­3p and its target ras protein activator like 2 (RASAL2) mRNA were determined by reverse transcription­quantitative polymerase chain reaction; protein expression was analyzed by western blotting. Plasmids expressing miR­654­3p mimics were constructed and transfected into A549 cells. In addition, the viability and apoptotic rate of cells were analyzed by an MTT assay and flow cytometry, respectively. A luciferase reporter assay was performed to verify whether RASAL2 is a target of miR­654­3p. Downregulated miR­654­3p and upregulated RASAL2 expression were observed in tumor tissues and cells. Cell viability was suppressed and the apoptotic rate was increased in the miR­654­3p mimics­transfected cells compared with the control. Luciferase activity was decreased in the RASAL2­3' untranslated region­wild type group treated with miR­654­3p mimics. Furthermore, the present study revealed that overexpression of miR­654­3p could suppress the viability and induce the apoptosis of cells by targeting RASAL2 in NSCLC. The present findings may contribute to developments in the treatment of NSCLC.


Assuntos
Apoptose , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Neoplasias Pulmonares/metabolismo , MicroRNAs/metabolismo , Proteínas de Neoplasias/metabolismo , RNA Neoplásico/metabolismo , Células A549 , Idoso , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Sobrevivência Celular , Feminino , Proteínas Ativadoras de GTPase/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , RNA Neoplásico/genética
4.
Oncol Rep ; 43(2): 395-404, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31894338

RESUMO

Lung cancer, which is a leading cause of cancer­related deaths, is diagnosed at a male to female ratio of 2.1:1. Serine­threonine kinase 31 (STK31) is a novel cancer/testis (CT)­related gene that is highly expressed in several types of cancers, such as lung and colorectal cancer, and plays crucial roles in cancer. In the present study, increased expression of STK31 and ß­catenin was observed in lung cancer tissues and cell lines. Downregulation of STK31 expression in lung cancer cells significantly inhibited their proliferation by arresting the cell cycle in the G1 phase concurrent with decreased ß­catenin, c­myc and cyclin D1 protein levels, while upregulation of STK31 had the opposite effects. In addition, STK31­induced lung cancer cell viability, proliferation, cell cycle progression, and expression of related genes were completely attenuated by a Wnt/ß­catenin inhibitor (XAV939). Similar to XAV939, a c­myc inhibitor (10058­F4) also significantly attenuated STK31­induced proliferation and cell cycle progression in lung cancer cells. Inhibiting c­myc and TRRAP significantly decreased the expression of STK31, and a chromatin immunoprecipitation (ChIP) assay confirmed that c­myc directly bound to the STK31 promoter. These results indicated that STK31 may act as an oncogene in lung cancer and that c­myc may be the transcription factor that promotes STK31 expression. Moreover, the results suggested that c­myc can also regulate STK31 expression in a positive feedback loop, and the downregulation of STK31 in lung cancer cells had an inhibitory effect on cell viability, cell proliferation and cell cycle progression, likely by inactivating the Wnt/ß­catenin pathway and positive feedback regulation by c­myc.


Assuntos
Neoplasias Pulmonares/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-myc/genética , beta Catenina/genética , Células A549 , Idoso , Idoso de 80 Anos ou mais , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Retroalimentação Fisiológica , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Masculino , Pessoa de Meia-Idade , Tiazóis/farmacologia , Regulação para Cima , Via de Sinalização Wnt
5.
Mol Med Rep ; 20(6): 5272-5278, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31638208

RESUMO

Dysregulation of microRNAs (miRNAs) is frequently observed during cancer development. Aberrant expression of miRNA­138­5p (miR­138­5p) has been found in many types of cancer. However, the role and the mechanisms underlying miR­138­5p function in non­small cell lung cancer (NSCLC) progression remain unknown. In the present study, miR­138­5p expression was identified to be decreased in tumor tissues compared with matched normal tissues from patients with NSCLC. In addition, low expression of miR­138­5p was detected in three NSCLC cell lines compared with a normal lung epithelium cell line. Moreover, CDK8 mRNA expression was increased in tumor tissues compared with matched normal tissues from patients with NSCLC, and an inverse association between miR­138­5p and CDK8 was observed. Furthermore, CDK8 was predicted to be a target of miR­138­5p. Dual luciferase assay confirmed that miR­138­5p could directly bind to the 3' untranslated region of CDK8 mRNA. In A549 cells, overexpression of miR­138­5p inhibited cell growth and significantly increased cell apoptosis rates and the number of cells in G0/G1 phase. Moreover, forced overexpression of CDK8 partially reversed miR­138­5p mimic­induced cell growth arrest and alteration of cell apoptosis and cell cycle. In conclusion, the present results suggested that miR­138­5p may be a tumor suppressor in NSCLC cells and a promising therapeutic target for treating patients with NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Quinase 8 Dependente de Ciclina/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , MicroRNAs/genética , Interferência de RNA , Regiões 3' não Traduzidas , Idoso , Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Humanos , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade
6.
Cancer Biomark ; 20(4): 597-608, 2017 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-28800319

RESUMO

Studies have shown that microRNAs (miRNAs) can promote or suppress tumor growth and therefore act as targets for cancer therapy. Hsa-miR-503-5p, a mature miRNA derived from 5' ends of pre-miR-503, has been proved to regulate cell proliferation, transformation, migration and invasion. However, the biological function of miR-503-3p derived from 3' ends of pre-miR-503 has never been reported. In current study, we found that miR-503-3p inhibits lung cancer cell viability and induces cell apoptosis. To better understand the molecular mechanism underlying the miR-503-3p participating in this process, PCR array and RNA-sequencing (RNA-seq) were performed and some differential expression genes were discovered between NC and miR-503-3p treated groups. Biological interaction network showed that p21 and CDK4 are the most important proteins involving miR-503-3p signal pathway. Dual-luciferase assay results shown miR-503-3p directly regulates the expression of p21 by targeting 3'-UTR of its mRNA. These results shed light on the potential roles of miR-503-3p, indicating that it may act as an anti-oncogene factor to inhibit lung cancer cell viability.


Assuntos
Apoptose/genética , Quinase 4 Dependente de Ciclina/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Interferência de RNA , Regiões 3' não Traduzidas , Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Expressão Gênica , Perfilação da Expressão Gênica/métodos , Genes Reporter , Humanos
7.
Cell Stress Chaperones ; 20(5): 805-10, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25981116

RESUMO

Cigarette smoking plays an important role in increased incidence of chronic obstructive pulmonary disease (COPD). The underlying mechanism in which cigarette smoking induced impairment of lung epithelial cells is still unknown. SIRT5 is a nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylase, which has been implicated in the regulation of metabolism, stress responses, and aging. Forkhead box O3 (FOXO3) belongs to the O subclass of the forkhead family of transcription factors. It is also involved in protection from oxidative stress by upregulating antioxidants in epithelial cells. Here, we show that cigarette smoke extract (CSE) induces SIRT5 to deacetylate FOXO3 at K271 and K290. Deacetylation of FOXO3 promotes its nuclear localization. Notably, transfection with FOXO3 K271R- or K290R-attenuated CSE-induced apoptosis in SIRT5 knocked down cells, suggesting the protective effects of SIRT5, is mediated by FOXO3. In contrast, CSE stress upregulates SIRT5, which activates FOXO3α leading to rescuing apoptosis. Thus, SIRT5 constitutes a determinant of apoptosis by CSE in lung epithelial cells.


Assuntos
Apoptose , Células Epiteliais , Fatores de Transcrição Forkhead/metabolismo , Sirtuínas/metabolismo , Fumar/efeitos adversos , Linhagem Celular , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Proteína Forkhead Box O3 , Humanos , Alvéolos Pulmonares/citologia , Doença Pulmonar Obstrutiva Crônica/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...