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










Base de dados
Intervalo de ano de publicação
1.
Tissue Eng Regen Med ; 21(4): 595-608, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38466361

RESUMO

BACKGROUND: The purpose of this study was to investigate the specific effects of signal transducer and activator of transcription 4 (STAT4)-induced long intergenic nonprotein coding RNA 1278 (LINC01278) on the growth of non-small cell lung cancer (NSCLC) cells involved in the microRNA (miR)-877-5p/activated transcription factor 4 (ATF4) axis. METHODS: NSCLC tumor tissue and adjacent normal tissue were collected. Human normal lung epithelial cell BEAS-2B and human NSCLC cell lines (H1299, H1975, A549, H2228) were collected. The expression levels of STAT4, LINC01278, miR-877-5p, and ATF4 were detected. A549 cells were screened for subsequent experiments. The proliferation ability of cells was detected by colony formation experiment. Cell apoptosis was tested by flow cytometry. Scratch test and transwell assay were used to detect the migration and invasion ability of cells. Biological function of LINC01278 in NSCLC was confirmed by xenograft experiments. RESULTS: Low expression miR-877-5p and high expression of STAT4, LINC01278 and ATF4 were detected in NSCLC. Silenced LINC01278 in A549 cell depressed cell proliferation, migration and invasion, but facilitated cell apoptosis. LINC01278 was positively correlated with STAT4 and could directly bind to miR-877-5p. Upregulating miR-877-5p suppressed NSCLC cell progression, while downregulating miR-877-5p had the opposite effect. Upregulating miR-877-5p abrogated the effects of silenced LINC01278 on NSCLC cell progression. MiR-877-5p targeted ATF4. ATF4 upregulation could partly restore the carcinogenic effect of LINC01278 in vitro and in vivo. CONCLUSION: Our data supports that STAT4-induced upregulation of LINC01278 promotes NSCLC progression by modulating the miR-877-5p/ATF4 axis, suggesting a novel direction for NSCLC treatment.


Assuntos
Fator 4 Ativador da Transcrição , Carcinoma Pulmonar de Células não Pequenas , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares , MicroRNAs , RNA Longo não Codificante , Fator de Transcrição STAT4 , Regulação para Cima , Humanos , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Fator 4 Ativador da Transcrição/metabolismo , Fator 4 Ativador da Transcrição/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/genética , Animais , Fator de Transcrição STAT4/metabolismo , Fator de Transcrição STAT4/genética , Invasividade Neoplásica , Linhagem Celular Tumoral , Células A549 , Camundongos Nus , Camundongos , Apoptose , Feminino , Masculino , Movimento Celular , Camundongos Endogâmicos BALB C
2.
ESC Heart Fail ; 8(4): 3214-3222, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34041859

RESUMO

AIMS: Myeloid differentiation protein 1 (MD1) was shown to ameliorate pressure overload-induced cardiac hypertrophy and fibrosis by negatively regulating the MEK-ERK1/2 and NF-κB pathways. However, whether MD1 modulates cardiac function and whether the Akt pathway mediates the benefits of MD1 in pressure overload-induced cardiac remodelling remain unclear. METHODS AND RESULTS: Male cardiac-specific transgenic MD1 (MD1-TG) mice, MD1-knockout (KO) mice and wild-type (WT) littermates aged 8-10 weeks were subjected to sham operation and aortic banding (AB) for 4 weeks. Then, left ventricular (LV) hypertrophy, fibrosis and function of the mice were assessed. When compared with WT-AB mice, MD1-TGs showed decreased cross-sectional area (CSA) of cardiomyocytes (P < 0.001), mRNA expression of ß-myosin heavy chain (ß-MHC) (P < 0.02), ratios of heart weight/body weight and heart weight/tibia length (P < 0.04) and collagen volume fraction (P < 0.001). The LV end-diastolic diameter was reduced, and LV ejection fraction and fractional shortening were improved in MD1-TG-AB mice than in WT-AB mice (P < 0.05). In cultured H9C2 cells, adenovirus vector-mediated MD1 overexpression decreased angiotensin II-induced mRNA expression of brain natriuretic peptide (BNP) and ß-MHC and cell CSA (P < 0.002), whereas knockdown of MD1 by shRNA exhibited opposite effects (P < 0.04). Mechanistically, MD1 suppressed pathological cardiac remodelling at least partly by blocking Akt pathway. Akt inactivation by MK2206 largely offset the pro-hypertrophic effects of MD1 deficiency in angiotensin II-stimulated cardiomyocytes. CONCLUSIONS: The Akt pathway mediates the protective effects of MD1 in pressure overload-induced cardiac remodelling in mice. Targeting MD1 may provide therapeutic strategy for the treatment of pathological cardiac remodelling and heart failure.


Assuntos
Proteínas Proto-Oncogênicas c-akt , Remodelação Ventricular , Animais , Diferenciação Celular , Hipertrofia Ventricular Esquerda , Masculino , Camundongos , Função Ventricular Esquerda
3.
Mol Med Rep ; 23(6)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33786635

RESUMO

Poor prognosis in patients with glioma is primarily due to rapid tumor growth and cell invasion and migration. In addition, microRNA (miR)­27b is decreased in metastatic glioma. The present study investigated whether sevoflurane inhibited glioma cell progression by targeting miR­27b. Cell proliferation was analyzed using a Cell Counting Kit­8 assay and a wound healing assay was used to detect cell migration. Western blotting and reverse transcription­quantitative PCR analysis were performed to determine the protein and mRNA expression levels. A dual luciferase assay was used to determine the relationship between vascular epithelial growth factor (VEGF) and miR­27b. VEGF was identified to be a direct target of miR­27b. Moreover, sevoflurane treatment increased the expression of miR­27b and decreased the expression of VEGF in U251 and U87 cells. Compared with the control group, sevoflurane inhibited the proliferation and migration of U251 and U87 cells, as well as the expression of matrix metalloproteinase (MMP)­2 and MMP­9, which were subsequently abolished by pre­treatment with an miR­27b inhibitor. The present results indicated the potential use of sevoflurane by anesthesiologists for the surgical resection of glioma, which may improve patient outcomes in the clinical setting.


Assuntos
Antineoplásicos/farmacologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Glioma/metabolismo , Sevoflurano/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
Biochem Biophys Res Commun ; 547: 15-22, 2021 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-33588234

RESUMO

BACKGROUND: Ciliary neurotrophic factor (CNTF), which is a neural peptide, has been reported to confer cardioprotective effects. However, whether CNTF-based gene therapy could prevent cardiac remodelling remains incompletely clear. In this study, we used adeno-associated viral vector serotype 9 (AAV9)-based cardiac gene therapy to test the effects of CNTF overexpression on adverse ventricular remodelling in angiotensin II (Ang II)-infused mice. METHODS: First, AAV9-EGFP and AAV9-CNTF constructs were generated with virus concentration at 5 × 1012 vg/ml. Next, postnatal (P3-P10) mice with C57BL/6J background were administered with 5 × 1011 vg of AAV9 recombinant genome diluted in 50 µl of saline, and delivered through intraperitoneal injection. Implantation of osmotic minipumps was performed in 8-week-old male mice and human Ang II solution was administrated in the mice subcutaneously for 14 days through the pumps. Finally, we evaluated the effects of CNTF overexpression on mouse cardiac function, hypertrophy and fibrosis, as well as investigated the possible mechanisms. RESULTS: Our data showed that CNTF overexpression in mouse cardiomyocytes prevents cardiac hypertrophy and fibrosis induced by chronic Ang II stimulation. Mechanistic study found that CNTF overexpression upregulated NFE2-related factor 2 (Nrf2) antioxidant pathway, coupled with decreased ROS level in the cardiac tissues. Additionally, inflammatory cytokines were found to be reduced upon cardiac CNTF overexpression in response to chronic Ang II stimulation. CONCLUSIONS: Altogether, these results provide further evidence that CNTF can alleviate the condition of cardiac remodelling induced by chronic Ang II stimulation. Therefore, our results suggest a potential therapeutic role of CNTF in cardiac pathological remodelling.


Assuntos
Cardiomegalia/prevenção & controle , Fator Neurotrófico Ciliar/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Remodelação Ventricular/fisiologia , Angiotensina II/administração & dosagem , Animais , Cardiomegalia/induzido quimicamente , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Modelos Animais de Doenças , Fibrose , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Vasoconstritores/administração & dosagem , Remodelação Ventricular/efeitos dos fármacos
5.
Open Med (Wars) ; 16(1): 104-116, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33506107

RESUMO

BACKGROUND: The aim of this study was to investigate the circ_0004370 expression in EC, its effects on cell proliferation, apoptosis, migration, invasion, and epithelial-mesenchymal transition (EMT) process, and the underlying regulatory mechanisms in EC. METHODS: The protein levels of COL1A1 and EMT-related proteins were detected by western blot. The role of circ_0004370 on cell viability, proliferation, and apoptosis was analyzed by Cell Counting Kit-8 (CCK-8) assay, colony formation assay, and flow cytometry, respectively. The transwell assay was used to examine cell migration and invasion. The binding sites between miR-1301-3p and circ_0004370 or COL1A1 were predicted by starbase software and confirmed by dual-luciferase reporter assay and RNA pull-down assay. RESULTS: We discovered that circ_0004370 was remarkably upregulated in EC tissues and cells. Knockdown of circ_0004370 inhibited cell proliferation, migration as well as invasion, and promoted apoptosis in vitro, while its effect was rescued by miR-1301-3p inhibition. And circ_0004370 mediated the EMT process in EC cells. Moreover, we explored its regulatory mechanism and found that circ_0004370 directly bound to miR-1301-3p and COL1A1 was verified as a target of miR-1301-3p. COL1A1 was highly expressed in EC cells and upregulation of COL1A1 reversed the effects of miR-1301-3p on cell proliferation, migration, invasion, and apoptosis. In addition, silencing of circ_0004370 reduced tumor volumes and weights in vivo. We showed that circ_0004370/miR-1301-3p/COL1A1 axis played the critical role in EC to regulate the cell activities. CONCLUSION: Circ_0004370 promotes EC proliferation, migration and invasion, and EMT process and suppresses apoptosis by regulating the miR-1301-3p/COL1A1 axis, indicating that circ_0004370 may be used as a potential therapeutic target for EC.

8.
Gene ; 726: 144136, 2020 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-31629817

RESUMO

Chronic systolic heart failure (CSHF) was a complex syndrome. Recently, vagus nerve stimulation (VNS), a novel treatment method, has emerged for the treatment of CSHF. therefore the aim of this study was to explore the possible mechanism of VNS treatment alleviating CSHF in rats. Firstly, we found after VNS treatment for 72 h, the level of B-type natriuretic peptide in VNS group was lower than that in CSHF group. In addition, VNS treatment induced the elevated left ventricular ejection fraction level, reduced left ventricular end diastolic volume and left ventricular end systolic volume level in VNS group, suggesting a mitigation of CSHF by VNS. Then we found the level of miR-183-3p in CSHF group was much lower than that in VNS group by High-throughput sequencing. The further results indicated that Bcl-2 interacting protein 3 like (BNIP3L) was identified as the target gene of miR-183-3p, and the expression of BNIP3L was notably reduced in rats of VNS group compared with CSHF group. Moreover, the down-regulated expression of miR-183-3p increased BNIP3L-mediated autophagy in rats of CSHF group compared with VNS group. Further mechanism findings demonstrated that up-regulation of miR-183-3p reduced the expression of BNIP3L, while down-regulation of miR-183-3p facilitated the expression of BNIP3L in H9c2 cells. miR-183-3p could also regulate autophagy by targeting BNIP3L in vitro, which was manifested by overexpression of miR-183-3p to inhibit BNIP3L-mediated autophagy. Our data demonstrated that VNS treatment benefited CSHF via the up-regulation of miRNA-183-3p, which reduced the BNIP3L-mediated autophagy, providing a new therapeutic direction for CSHF.


Assuntos
Autofagia/genética , Insuficiência Cardíaca Sistólica/genética , Proteínas de Membrana/genética , MicroRNAs/genética , Proteínas Proto-Oncogênicas/genética , Regulação para Cima/genética , Animais , Regulação para Baixo/genética , Masculino , Ratos , Ratos Wistar , Volume Sistólico/genética , Ativação Transcricional/genética , Estimulação do Nervo Vago/métodos , Função Ventricular Esquerda/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...