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.
Front Cell Dev Biol ; 10: 977960, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36407113

RESUMO

Purpose: Accumulating evidence suggests that solute carrier family 39 member 1 (SLC39A1) conceivably function as a tumor suppressor, but the underlying mechanism in renal cell carcinoma (RCC) is poorly understood. Methods: OSRC-2 renal cancer cells were first transfected with SLC39A1 overexpressed vectors and empty vectors and then used in transcriptomics, proteomics, and metabolomics integrated analyses. Results: SLC39A1 significantly altered several metabolisms at transcriptional, protein and metabolic levels, including purine and pyrimidine metabolism, amino acids and derivatives metabolism, lactose metabolism, and free fatty acid metabolism. Additionally, SLC39A1 could promote ferroptosis, and triggered significant crosstalk in PI3K-AKT signal pathway, cAMP signal pathway, and peroxisome proliferators-activated receptor (PPAR) signal pathway. Conclusion: We found SLC39A1 transfection impaired tumor metabolism and perturbed tumor metabolism-related pathways, which was a likely cause of the alteration in cell proliferation, migration, and cell cycle progression in RCC cells. These multi-omics analyses results provided both a macroscopic picture of molecular perturbation by SLC39A1 and novel insights into RCC tumorigenesis and development.

2.
J Hepatocell Carcinoma ; 9: 83-98, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35211427

RESUMO

BACKGROUND: Solute carrier family 39 member 1 (SLC39A1) has been identified as a zinc ion transport protein that possesses oncogenic properties in various types of cancers. However, its potential function in hepatocellular carcinoma (HCC) remains unknown. This study aimed to investigate the expression profile and potential mechanisms of SLC39A1 in HCC. METHODS: SLC39A1 expression was analyzed using multiple databases. The clinical significance and associated biological pathways of SLC39A1 were investigated using bioinformatics analysis. Potential correlations between SLC39A1 expression and tumor immunity in HCC were also evaluated using single-sample gene set enrichment analysis (GSEA). Sixty paired HCC samples were used to verify the expression pattern of SLC39A1. In vitro studies were performed to investigate the oncogenic effects of SLC39A1 in HCC. Western blot analysis was conducted to further investigate the possible involved signaling pathways. RESULTS: The overexpression of SLC39A1 in HCC was determined by bioinformatics analysis and was confirmed in tissues from our center. SLC39A1 overexpression was also significantly correlated with worse prognosis, advanced TNM stage, and histological grade. GSEA analysis demonstrated that SLC39A1 overexpression was involved in various tumor-related pathways, such as the cell cycle and Wnt signaling pathway. SLC39A1 knockdown repressed the proliferation, invasion, and migration abilities of HCC cells. Furthermore, SLC39A1 knockdown decreased the expression of the tumor progression-related proteins (eg, cyclin D1 and MMP2) and Wnt signaling pathway-related proteins (eg, Wnt3A and ß-catenin). In addition, SLC39A1 overexpression may be associated with impaired tumor immunity in HCC, as evidenced by the increased infiltration of Th2 cells and reduced infiltration of cytotoxic cells. CONCLUSION: These findings preliminarily suggested the crucial effect of SLC39A1 overexpression on HCC tumor progression and immunosuppression, suggesting its potential as a novel prognostic and therapeutic target in HCC.

3.
Bioengineered ; 12(1): 8147-8156, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34615436

RESUMO

Solute carrier family 39, member 1 (SLC39A1) is a member of the zinc-iron permease family and located to the cell membrane, acting as a zinc uptake transporter. However, the clinical impacts of SLC39A1 in early-stage hepatocellular carcinoma (EHCC) have not been defined. In this research, we compared the differential expression of SLC39A1 in EHCC and normal tissues based on tissue microarray, and the clinical significance of SLC39A1 in EHCC was evaluated as well. Compared with adjacent tissues, SLC39A1 was remarkably decreased in paired EHCC tissues. Besides, decreased SLC39A1 expression was significantly associated with several clinic-pathological features and serum biochemical indicators. Furthermore, Kaplan-Meier analysis exhibited that both overall survival (OS) and relapse-free survival (RFS) of patients with low expression of SLC39A1 were notably poorer than that of patients with high expression. Moreover, Cox regression analyses revealed that low expression of SLC39A1 was an independent prognostic factor for OS in patients with EHCC. Subgroup analysis also revealed beneficiary populations benefiting from the prognostic evaluation using SLC39A1 expression. Collectively, we summarized that downregulated expression of SLC39A1 is a worse prognostic factor for patients with EHCC, which can be used as a promising diagnostic and prognostic biomarker for EHCC.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/patologia , Proteínas de Transporte de Cátions/metabolismo , Regulação para Baixo , Neoplasias Hepáticas/patologia , Carcinoma Hepatocelular/metabolismo , Estudos de Casos e Controles , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Neoplasias Hepáticas/metabolismo , Masculino , Estadiamento de Neoplasias , Prognóstico , Análise de Sobrevida
4.
Cancer Cell Int ; 20(1): 573, 2020 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-33292262

RESUMO

BACKGROUND: Gliomas are one of the most common primary tumors of the central nervous system, and have an unfavorable prognosis. SLC39A1 is a zinc ion transport protein which inhibits the progression of prostate cancer. By studying the role and mechanism of SLC39A1 in the progression of gliomas, perhaps a new therapeutic target can be provided for their treatment. METHOD: The TCGA, CCGA, GSE16011, GSE44971 and GSE11260 data sets were employed to evaluate the expression level of SLC39A1 in paracancerous and glioma tissues. In addition, Kaplan-Meier analysis, Cox analysis, and the ESTIMATE and CIBERSORT algorithms were used to analyze its prognostic value and immune infiltration correlation. A CCK-8 and flow cytometer were used to measure the effects of SLC39A1 on U87 cell proliferation or apoptosis; RT-qPCR and western blot were used to detect its effects on the expression of MMP2\MMP9. RESULTS: SLC39A1 has up-regulated expression in glioma tissues. High SLC39A1 expression predicted significantly worse survival. Univariate and multivariate analysis show that SLC39A1 independently indicated poor prognosis in patients with gliomas. The expression of SLC39A1 is significantly correlated with clinical pathological parameters such as Grade, IDH mutation status, and 1p19q codeletion status. In vitro experimental results show that SLC39A1 promotes proliferation of glioma cells, inhibits their apoptosis, and promotes expression of MMP2\MMP9. In addition, it may affect infiltration of immune cells into the glioma microenvironment. CONCLUSION: SLC39A1 may serve as a new prognostic biomarker and potential target for treatment of gliomas.

5.
RNA Biol ; 11(11): 1355-63, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25692234

RESUMO

Osteoclasts are the exclusive cells of bone resorption. Abnormally activating osteoclasts can lead to low bone mineral density, which will cause osteopenia, osteoporosis, and other bone disorders. To date, the mechanism of how osteoclast precursors differentiate into mature osteoclasts remains elusive. MicroRNAs (miRNAs) are novel regulatory factors that play an important role in numerous cellular processes, including cell differentiation and apoptosis, by post-transcriptional regulation of genes. Recently, a number of studies have revealed that miRNAs participate in bone homeostasis, including osteoclastic bone resorption, which sheds light on the mechanisms underlying osteoclast differentiation. In this review, we highlight the miRNAs involved in regulating osteoclast differentiation and bone resorption, and their roles in osteoporosis.


Assuntos
Regulação da Expressão Gênica , MicroRNAs/genética , Osteoclastos/metabolismo , Osteoporose/genética , Reabsorção Óssea/genética , Osso e Ossos/metabolismo , Diferenciação Celular/genética , Redes Reguladoras de Genes , Homeostase/genética , Humanos , Proto-Oncogene Mas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA