Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Environ Toxicol ; 38(4): 809-819, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36620879

RESUMO

BACKGROUND: Colorectal cancer is a common malignant digestive tract tumor. This study aimed to explore the biological role and potential underlying mechanism of matrine in colorectal cancer. METHODS: The mRNA expression of AGRN was measured using RT-qPCR. Cell proliferation, migration, invasion and apoptosis were determined using CCK-8, EdU, transwell assays and flow cytometry, respectively. Xenograft tumor experiment was performed to explore the action of matrine and AGRN on tumor growth in colorectal cancer in vivo. Immunohistochemistry (IHC) assay was applied for AGRN, ß-catenin, and c-Myc expression in the tumor tissues from mice. RESULTS: Matrine dramatically repressed cell growth and reduced the level of AGRN in colorectal cancer cells. AGRN expression was boosted colorectal cancer tissues and cells. AGRN downregulation depressed cell proliferation, migration, invasion, and enhanced cell apoptosis in colorectal cancer cells. Moreover, matrine showed the anti-tumor effects on colorectal cancer cells via regulating AGRN expression. AGRN knockdown could inactivate the Wnt/ß-catenin pathway in colorectal cancer cells. We found that AGRN downregulation exhibited the inhibition action in the progression of colorectal cancer by modulating the Wnt/ß-catenin pathway. In addition, matrine could inhibit the activation of the Wnt/ß-catenin pathway through regulating AGRN in colorectal cancer cells. Furthermore, xenograft tumor experiment revealed that matrine treatment or AGRN knockdown repressed the development of colorectal cancer via the Wnt/ß-catenin pathway in vivo. CONCLUSION: Matrine retarded colorectal cancer development by modulating AGRN to inactivate the Wnt/ß-catenin pathway.


Assuntos
Neoplasias Colorretais , Matrinas , Humanos , Animais , Camundongos , beta Catenina/metabolismo , Via de Sinalização Wnt , Regulação para Baixo , Apoptose/genética , Neoplasias Colorretais/genética , Proliferação de Células/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica
2.
Mol Cell Biochem ; 477(11): 2669-2679, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35604518

RESUMO

Nuclear receptor subfamily 3 group c member 2 (NR3C2) has been reported to function as a tumor suppressor in several tumors. However, the clinical significance and potential action mechanisms of NR3C2 in colon cancer (COAD) remain unclear. NR3C2 expression and its correlation with clinicopathological features in COAD were analyzed based on the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Receiver operating characteristic (ROC) curves and Human Protein Atlas (HPA) database were used to evaluate the diagnostic and prognostic values of NR3C2 in COAD. Immune infiltration and DNA methylation analyses were performed by Gene Set Cancer Analysis (GSCA) database. NR3C2-correlated genes were identified by UALCAN database and subjected to gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment pathway analyses. Cell apoptosis and proliferation were evaluated using TUNEL and CCK-8 assays, respectively. NR3C2 was downregulated in COAD based on TCGA and GEO databases, which may be due to promoter hypermethylation. NR3C2 expression was correlated with prognosis and immune infiltration of COAD. High NR3C2 expression displayed good diagnostic value in COAD. KEGG pathway analysis presented that NR3C2-correlated genes were mainly clustered in choline metabolism in cancer and apoptosis. In vitro experiments confirmed that NR3C2 overexpression induced apoptosis and suppressed proliferation in COAD cells. In conclusion, our study revealed the potential prognostic and diagnostic values of NR3C2 and provided insights into understanding the tumor-suppressive role of NR3C2 in COAD progression.


Assuntos
Neoplasias do Colo , Metilação de DNA , Humanos , Neoplasias do Colo/metabolismo , Regiões Promotoras Genéticas , Receptores de Mineralocorticoides/metabolismo
3.
Environ Toxicol ; 37(3): 435-445, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34773443

RESUMO

Colorectal cancer (CRC) is one of the prevalent types of human malignancies and ranks as the second leading cause of cancer-associated death worldwide. Dysregulated miRNAs have been promulgated as oncogenes or tumor-suppressive genes participating in the initiation and progression of CRC. A recent study reported that miR-346 was highly expressed in CRC patients. However, the biological role and underlying mechanism of miR-346 in CRC remain elusive. qRT-PCR and western blot assays were employed to detect miR-346 and LIM homeobox domain 6 (LHX6) expression in CRC cells. Cell proliferation was evaluated by CCK-8 and BrdU assays. Apoptosis was evaluated by TUNEL assay. The interaction between miR-346 and LHX6 was assessed by luciferase reporter assay. Results showed that miR-346 expression was increased and LHX6 expression was reduced in CRC cells. miR-346 knockdown and LHX6 overexpression inhibited proliferation and promoted apoptosis of CRC cells. Additionally, we found that miR-346 negatively regulated LHX6 expression in CRC cells by directly targeting LHX6. LHX6 knockdown partially attenuated anti-miR-346-induced proliferation reduction and apoptosis promotion in CRC cells. Furthermore, miR-346 knockdown inhibited the protein kinase B (Akt)/mechanistic target of rapamycin (mTOR) pathway in CRC cells by targeting LHX6. The present study indicated that miR-346 knockdown repressed cell growth in CRC cells by upregulating LHX6, and this was associated with inactivation of the Akt/mTOR pathway.


Assuntos
Neoplasias Colorretais , MicroRNAs , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas com Homeodomínio LIM/genética , MicroRNAs/genética , Proteínas do Tecido Nervoso/genética , Fatores de Transcrição/genética , Regulação para Cima
4.
Life Sci ; 232: 116679, 2019 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-31340168

RESUMO

AIMS: Amplified in liver cancer 1 gene (ALC1), a recently identified oncogene, was reported to be overexpressed in esophageal cancer cell lines and identified as a target oncogene in esophageal cancer pathogenesis. However, little literature is available to illustrate its significance in cisplatin resistance of esophageal squamous cell carcinoma (ESCC) cells. The aim of the current study was to investigate the effect of ALC1 on cisplatin cytotoxicity of ESCC cells and to study the potential mechanisms. MAIN METHODS: ALC1 at mRNA and protein levels were detected by qRT-PCR and western blot, respectively. Cell viability was evaluated using CCK-8 assay. Apoptosis was assessed using caspase-3/7 activity assay and flow cytometry analysis. Glycolysis level was evaluated by measuring glucose consumption and lactate production. The protein levels of p-protein kinase B (Akt) and Akt were determined by western blot. KEY FINDINGS: ALC1 was highly expressed in ESCC cells compared with human normal esophageal epithelial Het-1A cells. ALC1 knockdown suppressed the viability, induced apoptosis and enhanced cisplatin cytotoxicity in ESCC cells. In addition, ALC1 knockdown inhibited glycolysis and inactivated the phosphatidylinositol 3-kinase (PI3K)/Akt pathway in ESCC cells. Mechanistically, activation of the PI3K/Akt pathway by 740Y-P blocked the effects of ALC1 knockdown on cisplatin cytotoxicity and glycolysis in ESCC cells. In contrast, inhibition of the PI3K/Akt pathway by LY294002 or glycolysis by 2-deoxyglucose resisted the effect of ALC1 overexpression on cisplatin cytotoxicity in ESCC cells. SIGNIFICANCE: ALC1 knockdown enhanced cisplatin cytotoxicity of ESCC cells by inhibition of glycolysis through inactivation of the PI3K/Akt pathway.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/patologia , Cisplatino/farmacologia , DNA Helicases/genética , Proteínas de Ligação a DNA/genética , Neoplasias Esofágicas/patologia , Técnicas de Silenciamento de Genes , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Apoptose , Carcinoma de Células Escamosas/enzimologia , Carcinoma de Células Escamosas/metabolismo , Ativação Enzimática , Neoplasias Esofágicas/enzimologia , Neoplasias Esofágicas/metabolismo , Glicólise , Humanos , Células Tumorais Cultivadas
5.
J Cell Biochem ; 119(6): 4783-4791, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29331036

RESUMO

Many studies have shown that randomized clinical trial with long-term follow-up found no improvement in stage II esophageal carcinoma (EC) patients receiving preoperative neoadjuvant chemoradiotherapy or chemotherapy treatment, this limitation underscored the urgent need for novel and reliable biomarkers for prognosis and prediction in stage II EC. miR-21 is frequently over-expressed while programmed cell death 4 (PDCD4) is often down-regulated in solid tumors. This study aimed to investigate the clinicopathological and prognostic significance of miR-21 and PDCD4 expression and to elucidate any correlation between miR-21 and PDCD4 expression in stage II EC patients. The expression level of miR-21 was up-regulated while the PDCD4 protein was down-regulated in stage II EC tissues compared with the adjacent non-cancerous tissues. Analyses of the clinicopathological parameters indicated that miR-21 expression was associated with differentiation grade, T stage, and N stage. PDCD4 protein expression was associated with T stage, N stage, and tumor size. The univariate linear regression analysis suggested a significant negative correlation between miR-21 and PDCD4 expression. The Kaplan-Meier curve showed that high miR-21 expression or low PDCD4 expression predicted poor progression-free survival (PFS) and overall survival (OS) of patients with stag II EC. In conclusion, both up-regulated miR-21 and down-regulated PDCD4 expression were associated with the aggressive progression and poor prognosis of stage II EC. miR-21 and PDCD4 might be potential biomarkers of tumor progression and indicators of prognosis of stag II EC.


Assuntos
Proteínas Reguladoras de Apoptose/biossíntese , Neoplasias Esofágicas , Regulação Neoplásica da Expressão Gênica , MicroRNAs/biossíntese , Proteínas de Neoplasias/biossíntese , RNA Neoplásico/biossíntese , Proteínas de Ligação a RNA/biossíntese , Idoso , Intervalo Livre de Doença , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/mortalidade , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/cirurgia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Taxa de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...