Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Healthc Eng ; 2022: 6556208, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35494527

RESUMEN

Background: Chronic obstructive pulmonary disease (COPD) is one of the most common chronic respiratory diseases in the world. COPD is a general term for a class of lung diseases, including emphysema, chronic bronchitis, and refractory asthma. It is characterized by irreversible airflow obstruction and chronic tracheal inflammation. Objective: This study aimed to investigate the expression and related mechanisms of miR-100 and TRIB2 in patients with COPD. Methods: We collected the serum of patients admitted to our hospital and healthy volunteers undergoing physical examination at the same time, pulmonary fibroblasts were purchased for the experiments, miR-100 was overexpressed, and TRIB2 expression was inhibited in cells. The miR-100 and TRIB2 expression levels in serum and cells were detected by qRT-PCR and Western blot, cell proliferation and apoptosis were detected by CCK-8 and flow cytometry, and the relationship between miR-100 and TRIB2 was explored by the dual-luciferase report. Results: The miR-100 expression in the serum of the COPD group was expressed normally, while the TRIB2 expression was expressed abnormally (p < 0.05). The AUC of serum miR-146a and TRIB2 for COPD diagnosis were 0.965 and 0.954, respectively. Overexpressing miR-100 and inhibiting the TRIB2 expression could decrease cell proliferation and increase apoptosis rate. According to the dual-luciferase report, miR-100 and TRIB2 had a targeted regulatory relationship. Rescue experiments showed that overexpressing TRIB2 could reverse the changes of cell proliferation and apoptosis caused by overexpression of miR-100. Conclusion: miR-100 and TRIB2 were expressed abnormally in serum of COPD patients, and miR-100 could inhibit proliferation of pulmonary fibroblasts and promote their apoptosis.


Asunto(s)
MicroARNs , Enfermedad Pulmonar Obstructiva Crónica , Apoptosis , Proteínas Quinasas Dependientes de Calcio-Calmodulina , Proliferación Celular , Humanos , Inflamación , MicroARNs/genética , MicroARNs/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/genética
2.
Bioengineered ; 12(2): 11959-11977, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34818974

RESUMEN

MicroRNA (miR)-140-3p has been proved to repress lung adenocarcinoma (LUAD), and our study aims to further evaluate the mechanism. Bioinformatic analyses were performed. The viability, proliferation, migration, invasion and angiogenesis of transfected LUAD cells were all determined via Cell Counting Kit-8, colony formation, Scratch, Transwell, and tube formation assays. The targeting relationship between miR-140-3p and thymidylate synthetase (TYMS) was confirmed by dual-luciferase reporter assay. Relative expressions of miR-140-3p, TYMS, epithelial-to-mesenchymal transition- (E-cadherin, N-cadherin, vimentin), angiogenesis- (vascular endothelial growth factor (VEGF)), and apoptosis-related factors (cleaved caspase-3, B-cell lymphoma-2 (Bcl-2), Bcl-2-associated X protein (Bax)) were quantified by quantitative real-time polymerase chain reaction or Western blot. TYMS was high-expressed yet miR-140-3p was low-expressed in LUAD cells. Upregulation of miR-140-3p inhibited TYMS expression, viability, colony formation, migration, invasion, and tube length within LUAD cells, while downregulation of miR-140-3p did oppositely. Silenced TYMS, the downstream target gene of miR-140-3p, reversed the effects of miR-140-3p downregulation on TYMS expression, cell viability, colony formation, migration, invasion, and tube length as well as the metastasis-, apoptosis- and angiogenesis-related proteins in LUAD cells. Upregulation of miR-140-3p inhibited the proliferation, migration, invasion and angiogenesis of LUAD cells via targeting TYMS.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Movimiento Celular/genética , Neoplasias Pulmonares/genética , MicroARNs/metabolismo , Neovascularización Patológica/genética , Timidilato Sintasa/metabolismo , Adenocarcinoma del Pulmón/patología , Apoptosis/genética , Secuencia de Bases , Línea Celular Tumoral , Proliferación Celular/genética , Supervivencia Celular/genética , Regulación hacia Abajo/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Neoplasias Pulmonares/patología , MicroARNs/genética , Invasividad Neoplásica , Timidilato Sintasa/genética , Regulación hacia Arriba/genética
3.
J Cancer Res Clin Oncol ; 144(4): 667-674, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29372377

RESUMEN

OBJECTIVE: Circular RNAs (circRNAs) are a novel class of non-protein-coding RNA. Emerging evidence indicates that circRNAs participate in the regulation of many pathophysiological processes. This study aims to explore the expression profiles and pathological effects of circRNAs in non-small cell lung cancer (NSCLC). METHODS: Human circRNAs microarray analysis was performed to screen the expression profile of circRNAs in NSCLC tissue. Expressions of circRNA and miRNA in NSCLC tissues and cells were quantified by qRTPCR. Functional experiments were performed to investigate the biological functions of circRNA, including CCK-8 assay, colony formation assay, transwell assay and xenograft in vivo assay. RESULTS: Human circRNAs microarray revealed a total 957 abnormally expressed circRNAs (> twofold, P < 0.05) in NSCLC tissue compared with adjacent normal tissue. In further studies, hsa_circ_0007385 was significantly up regulated in NSCLC tissue and cells. In vitro experiments with hsa_circ_0007385 knockdown resulted in significant suppression of the proliferation, migration and invasion of NSCLC cells. In vivo xenograft assay using hsa_circ_0007385 knockdown, significantly reduced tumor growth. Bioinformatics analysis and luciferase reporter assay verified the potential target miR-181, suggesting a possible regulatory pathway for hsa_circ_0007385. CONCLUSION: In summary, results suggest hsa_circ_0007385 plays a role in NSCLC tumorigenesis, providing a potential therapeutic target for NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , ARN/genética , Células A549 , Animales , Carcinogénesis/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular/genética , Técnicas de Silenciamiento del Gen , Células HEK293 , Xenoinjertos , Humanos , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Análisis por Micromatrices , Oncogenes , ARN Circular
4.
Biochem Biophys Res Commun ; 495(1): 20-26, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29079188

RESUMEN

Pulmonary fibrosis (PF) is a chronic, fibrosing interstitial pneumonia and devastating disease. Here we investigated the potential roles of Kruppel-like factor 2 (KLF2) on pulmonary fibrosis and inflammation response. A mouse model of pulmonary fibrosis was established by intratracheal injection of bleomycin (BLM). The mRNA and protein levels of KLF2 were assayed by RT-PCR and Western blotting respectively. The extent of lung fibrosis was determined using hematoxylin and eosin (HE) staining and Masson's trichrome staining, and the hydroxyproline content was quantified. RT-PCR was used to evaluate the mRNA expression of collagen type 1a1 (col1a1), col3a1, α-SMA, TNF-α, IL-1ß and IL-6. The concentrations of TNF-α, IL-1ß, and IL-6 in bronchoalveolar lavage fluid (BALF) and lung tissue were examined by ELISA. Also, the effects of KLF2 on activator protein-1 (AP-1) were evaluated by measuring the c-Jun and c-Fos protein levels. We found that KLF2 was remarkably downregulated in BLM-treated rats, both in mRNA and protein levels. Additionally, overexpression of KLF2 attenuated the destruction of the alveolar space and pulmonary interstitial collagen hyperplasia, and deposition reduced the expression of col1a1, col3a1, and α-SMA, and blocked the production of TNF-α, IL-1ß, and IL-6 in BALF and lung tissue in vivo. Moreover, adenoviral transduction of KLF2 inhibited TGF-ß1-induced expression of col1a1, col3a1, and α-SMA in vitro. Mechanically, BLM up-regulated c-Jun and c-Fos expression, which was impeded by KLF2 overexpression. Taken together, our data indicate that KLF2 attenuates pulmonary fibrosis and inflammation, possibly through the regulation of AP-1.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/metabolismo , Fibrosis Pulmonar/metabolismo , Factor de Transcripción AP-1/metabolismo , Actinas/genética , Animales , Bleomicina/toxicidad , Línea Celular , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Colágeno Tipo III/genética , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Femenino , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/metabolismo
5.
J Cell Biochem ; 119(2): 1746-1754, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28786504

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a chronic airway inflammation and its exacerbation is often accompanied by Aspergillus fumigatus (A. fumigatus) infection. Increasing evidences demonstrated the potent antioxidant and -inflammatory effects of crocin. However, the role of crocin in A. fumigatus-induced inflammation is still unknown. We aimed to evaluate the role of crocin in inflammation response induced by A. fumigatus in human bronchial epithelial cells and the possible mechanisms. BEAS-2B and NHBE cells were pretreated with crocin for 24 h, and then A. fumigatus conidia were added for 24 h. A. fumigatus treatment exhibited a significant higher TNF-α, IL-8, IL-6, and IL-1ß level (P < 0.05), whereas crocin pretreatment significantly inhibited A. fumigatus induced the pro-inflammatory cytokines (P < 0.05). NF-κB inhibitor PDTC inhibited pro-inflammatory cytokines release triggered by A. fumigatus (P < 0.05). Furthermore, crocin suppressed A. fumigatus induced NF-κB p65 nuclear translocation, the phosphorylation of IKKα and IκBα, the degradation of IκBα and NF-κB reporter activity. Crocin pretreatment also resulted in an inhibition of A.fumigatus-induced ROS production (P < 0.05). Taken together, these results indicate that crocin may prevent A. fumigatus-induced inflammation through suppressing NF-κB signal pathway.


Asunto(s)
Antiinflamatorios/farmacología , Aspergillus fumigatus/efectos de los fármacos , Carotenoides/farmacología , FN-kappa B/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Línea Celular , Células Epiteliales/inmunología , Humanos , Quinasa I-kappa B/metabolismo , Modelos Biológicos , Inhibidor NF-kappaB alfa/metabolismo , Prolina/análogos & derivados , Prolina/farmacología , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/microbiología , Transducción de Señal/efectos de los fármacos , Tiocarbamatos/farmacología , Factor de Transcripción ReIA/metabolismo
6.
Oncol Lett ; 14(1): 733-736, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28693227

RESUMEN

We studied the relationship between the polymorphisms of -800G/A and +915G/C in transforming growth factor-ß1 (TGF-ß1) gene and lung cancer susceptibility. The sequence-specific primer polymerase chain reaction (PCR-SSP) technique was used to test 156 non-small cell lung cancer (NSCLC) patients that were selected as the observation group and 156 patients with pneumonia and tuberculosis that were selected as the control group (age and gender 1:1 proximal matching principle) and the polymorphisms of the first exon -800G/A and +915G/C TGF-ß1 genes. The expression of TGF-ß1 levels in peripheral blood was detected using ELISA. The proportion of -800G/A gene AA subtype and A allelic gene in the observation group was significantly higher than that in the control group, while the proportion of +915G/C gene CC subtype and C allelic gene was also significantly higher than that in the control group (P<0.05). The cancer risk [odds ratio (OR)] of patients with A allelic gene in -800G/A gene was 4.8 (95% CI=2.563-6.537, P<0.05), while the cancer risk (OR) of patients with C allelic gene in +915G/C gene was 4.7 (95% CI=2.317-5.864, P<0.05). The serum TGF-ß1 expression levels of -800G/A gene AA subtype in the observation group was significantly higher than the GG type, GA type and the control group, while the TGF-ß1 level of +915G/C gene CC subtype was significantly higher than the GG type, GC type and the control group (P<0.05). Therefore, the polymorphisms of -800G/A and +915G/C in TGF-ß1 gene are closely related to the lung cancer susceptibility.

7.
Exp Physiol ; 102(5): 598-606, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28295786

RESUMEN

NEW FINDINGS: What is the central question of this study? Airway angiogenesis occurs in asthma, and airway smooth muscle (ASM) cells have been reported to be capable of promoting airway angiogenesis. What is the potential mechanism by which ASM cells harvested from patients with asthma are capable of promoting airway angiogenesis? What is the main finding and its importance? Endogenous STAT3 mediated the pro-angiogenic ability of ASM cells by directly activating VEGF signalling. These findings contribute to the understanding of airway angiogenesis in pathology and could represent a possible therapeutic target for asthma. Airway angiogenesis indicates the specific vascular structure remodelling that occurs in asthma. Airway smooth muscle (ASM) cells have been reported to be capable of promoting airway angiogenesis; however, the potential mechanism is not yet fully defined. Herein, we investigated the role of signal transducer and activator of transcription 3 (STAT3) in the progress of airway angiogenesis. Western blot analysis showed that STAT3 activation was aberrantly upregulated in ASM tissues of patients with asthma and ASM cells that were exposed to cytokines to imitate the airway conditions in patients with asthma. Compared with the control group, both the inhibition of STAT3 activation and the silencing of endogenous STAT3 in ASM cells significantly reduced the proliferation, migration and tube-forming ability of human lung microvascular endothelial cells induced by the conditioned medium (CM) of ASM cells. The increased proliferation and migration of human aortic vascular smooth muscle cells were also repressed by inhibition of STAT3 in ASM cells. Besides, the increased activity of VEGF signalling was observed in ASM cells and the CM by RT-PCR and Western blotting assay, whereas this increased activity was reduced by STAT3 silencing. Further studies indicated that STAT3 regulated VEGF activation by directly interacting with the binding site on the 5' region of the VEGF gene. The increase in STAT3-induced pro-angiogenic activity of ASM cells was significantly decreased by administration of VEGF neutralizing antibody. In conclusion, we provided evidence that endogenous STAT3 mediates the pro-angiogenic ability of ASM cells by directly activating VEGF signalling, which could represent a possible therapeutic target for asthma.


Asunto(s)
Músculo Liso/metabolismo , Músculo Liso/fisiología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/fisiología , Neovascularización Fisiológica/fisiología , Factor de Transcripción STAT3/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Aorta/metabolismo , Aorta/fisiopatología , Asma/metabolismo , Asma/fisiopatología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Humanos , Pulmón/metabolismo , Pulmón/fisiopatología , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología
8.
Mol Cell Biochem ; 432(1-2): 91-98, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28316062

RESUMEN

Claudin-1 (CLDN-1) is one of main tight junction components that play an important role in epithelial-mesenchymal transition (EMT). However, the effects of CLDN-1 on the migration and EMT induced by TGF-ß1 in primary normal human bronchial epithelial (NHBE) and BEAS-2B cells have not been clear. The expression of CLDN-1 was quantified by Western blotting in NHBE and BEAS-2B cells. Cell migration and invasion were detected using transwell assays. The expression level of E-cadherin, N-cadherin, α-SMA, and Vimentin was evaluated by quantitative real-time PCR and Western blotting. Here we showed that the protein expression of CLDN-1 was increased exposed to TGF-ß1 in a dose- and time-dependent manner. Knockdown of CLDN-1 using small interfering CLDN-1 RNA (siCLDN-1) prevented the migration and invasion in NHBE and BEAS-2B cells. Moreover, depletion of CLDN-1 promoted the E-cadherin expression and decreased the mRNA and protein levels of N-cadherin, α-SMA, and Vimentin induced by TGF-ß1. Furthermore, CLDN-1 silencing resulted in the reduction of the Notch intracellular domain (NICD) and hairy enhancer of split-1 (Hes-1) in mRNA and protein level. Jagged-1, an activator of Notch signaling pathway, abrogated the protective function of siCLDN-1 in migration and EMT. In conclusion, CLDN-1 promoted the migration and EMT through the Notch signaling pathway.


Asunto(s)
Bronquios/metabolismo , Movimiento Celular , Claudina-1/metabolismo , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Receptores Notch/metabolismo , Transducción de Señal , Bronquios/citología , Línea Celular , Claudina-1/genética , Células Epiteliales/citología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...