Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Tissue Cell ; 83: 102128, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37413858

RESUMO

AIM: This study aims to explore the possible effect of Astragaloside IV (AS-IV) on necrotizing enterocolitis (NEC) neonatal rat models and verify the possible implication of TNF-like ligand 1 A (TL1A) and NF-κB signal pathway. METHODS: NEC neonatal rat models were established through formula feeding, cold/asphyxia stress and Lipopolysaccharide (LPS) gavage method. The appearance, activity and skin as well as the pathological status of rats subjected to NEC modeling were assessed. The intestinal tissues were observed after H&E staining. The expression of oxidative stress biomarkers (SOD, MDA and GSH-Px) and inflammatory cytokines (TNF-α, IL-1ß and IL-6) were detected by ELISA and qRT-PCR. Western blotting and immunohistochemistry were applied to detect expressions of TL1A and NF-κB signal pathway-related proteins. Cell apoptosis was assessed by TUNEL. RESULTS: NEC neonatal rat models were established successfully, in which TL1A was highly expressed and NF-κB signal pathway was activated, while TL1A and NF-κB signal pathway can be suppressed by AS-IV treatment in NEC rats. Meanwhile, inflammatory response in intestinal tissues was increased in NEC rat models and AS-IV can attenuate inflammatory response in NEC rats through inhibiting TL1A and NF-κb signal pathway. CONCLUSION: AS-IV can inhibit TL1A expression and NF-κb signal pathway to attenuate the inflammatory response in NEC neonatal rat models.


Assuntos
Enterocolite Necrosante , NF-kappa B , Ratos , Animais , NF-kappa B/metabolismo , Animais Recém-Nascidos , Enterocolite Necrosante/tratamento farmacológico , Enterocolite Necrosante/metabolismo , Enterocolite Necrosante/patologia , Ligantes , Ratos Sprague-Dawley , Transdução de Sinais , Inflamação/patologia , Modelos Animais de Doenças
2.
Biochem Genet ; 61(2): 538-550, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35984538

RESUMO

Glioblastoma (GBM) is the most malignant and challenging type of astrocytoma and also notoriously acknowledged as the most common primary brain tumor globally. Currently, chemotherapy is the most master therapy for tumor and is essential in clinical treatment for GBM. Nevertheless, the characterization of chemotherapy resistance seriously hinders clinical chemotherapy treatment. Accordingly, there are imperious demands for the exploitation of novel chemosensitizer to promote the efficacy of chemotherapy. Our current study was conducted to probe into the potential impacts of microRNA (miR)-640 on the chemosensitivity in GBM and the associated underlying mechanism. Initially, TargetScan software was utilized to predict the targeted genes of miR-640, and the target relationship between miR-640 and Bcl-2-modifying factor (BMF) was validated by double luciferase report assay. Additionally, to explore the role of miR-640/BMF in U251 cells, miR-640 inhibitor/BMF-siRNA was used. U251 cells were processed with 100 µM temozolomide (TMZ) and detected with CCK-8 kit. Eventually, RT-qPCR and Western blotting were used for evaluating Bcl-2, Bax mRNA, and protein expression level. Flow cytometry analysis was performed to measure cellular apoptosis. Initially, the results indicated that BMF was the target gene of miR-640. MiR-640 negatively regulated BMF expression in GBM cells. Besides, the findings revealed that miR-640 inhibition significantly inhibited U251 cell proliferation, promoted cell apoptosis, and increased the sensitivity of GBM cells to TMZ by targeting BMF. Moreover, BMF overexpression significantly suppressed U251 cell proliferation, induced cell apoptosis, and increased the sensitivity of GBM cells to TMZ. Inhibition of miR-640 expression enhances chemosensitivity of human GBM cells to TMZ by targeting BMF.


Assuntos
Neoplasias Encefálicas , Glioblastoma , MicroRNAs , Humanos , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , MicroRNAs/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Resistencia a Medicamentos Antineoplásicos/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Apoptose/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Regulação Neoplásica da Expressão Gênica
3.
Exp Ther Med ; 24(3): 578, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35949317

RESUMO

Mycoplasma pneumoniae (M. pneumoniae) is a contributing factor to community-acquired pneumonia in children. The present study sought to explain the underlying mechanism of azithromycin (AZM) combined with methylprednisolone (MP) in the treatment of M. pneumoniae infection. Peripheral blood samples were obtained from patients with M. pneumoniae and healthy volunteers for analysis. A549 cells were infected with M. pneumoniae to construct an in vitro cell model with M. pneumoniae, followed by treatment with AZM and MP. Cell Counting Kit-8 and TUNEL assays were conducted to detect cell viability and apoptosis. RT-qPCR was employed to measure the expression levels of microRNA (miR)-499a-5p and STAT3. Western blotting was performed to measure the expression of STAT3 and apoptosis-related proteins. Luciferase report assay was performed to verify the binding site between miR-499a-5p and STAT3. The production of inflammatory cytokines was determined using ELISA kits. The results exhibited the downregulated miR-499a-5p and dysregulated inflammatory cytokines in peripheral blood of patients and M. pneumoniae-infected A549 cells. AZM and MP treatment alone or combined significantly inhibited inflammatory response, cell viability loss and promoted apoptosis in A549 cells infected with M. pneumoniae, which was partly reversed by inhibition of miR-499a-5p. Furthermore, miR-499a-5p could negatively regulate its direct target STAT3. In addition, STAT3 is also regulated by AZM and MP. Collectively, the present results suggested that combination treatment of AZM and MP could inhibit M. pneumoniae infection-induced inflammation, cell viability loss and promoted apoptosis partly by regulating miR-499a-5p/STAT3 axis.

4.
Front Oncol ; 11: 638148, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33791223

RESUMO

In recent years, the consumption of over-the-counter probiotics to promote health has grown rapidly worldwide and become an independent industry. In medicine, various studies have demonstrated that probiotics can help improve the immune system and intestinal health. They are usually safe, but in some rare cases, they may cause concerning adverse reactions. Although the use of probiotics has been widely popularized in the public, the results of many probiotic clinical trials are contradictory. Particularly in cancer patients, the feasibility of probiotic management providing benefits by targeting cancer and lessening anticancer side effects requires further investigation. This review summarizes the interactions between probiotics and the host as well as current knowledge on the pros and cons of utilizing probiotics in cancer patients.

5.
Oncol Lett ; 21(2): 161, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33552279

RESUMO

The effects of microRNAs (miRNAs/miRs) on glioblastoma have attracted the attention of researchers in the last 7 years. However, the role of miR-640 and its targeted gene, Slit guidance ligand 1 (SLIT1), in the development of glioblastoma are not yet fully understood. The present study aimed to investigate the role of miR-640 in the proliferation and adhesion of glioblastoma. Reverse transcription-quantitative PCR analysis was performed to detect miR-640 and SLIT1 expression in glioblastoma tissues and cells. In addition, the Dual-luciferase reporter and RNA-pull down assays were performed to assess the association between miR-640 and SLIT1. The Cell Counting Kit-8, BrdU ELISA, cell adhesion and caspase-3 activity assays were also performed to assess cell viability, proliferation, adhesion and apoptosis of glioblastoma cells, respectively. The results demonstrated that miR-640 expression was upregulated in glioblastoma tissues and cells. In addition, miR-640 promoted the cell viability, proliferation and adhesion of glioblastoma cells, while inhibiting cell apoptosis. SLIT1, a direct downstream target of miR-640, was demonstrated to be downregulated in glioblastoma tissues and cells. Furthermore, overexpression of SLIT1 attenuated the promotive effect of miR-640 on glioblastoma cells. Taken together, these results suggest that miR-640 accelerates the proliferation and adhesion of glioblastoma cell lines by targeting and suppressing SLIT1.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...