Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Cell Commun Signal ; 20(1): 92, 2022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35715860

RESUMO

BACKGROUND: Triple-negative breast cancer (TNBC) is a highly aggressive subtype of breast cancer with poor prognosis and limited treatment. As a major component of the tumor microenvironment, tumor-associated macrophages (TAMs) play an important role in facilitating the aggressive behavior of TNBC. This study aimed to explore the novel mechanism of TAMs in the regulation of epithelial-mesenchymal transition (EMT) and cancer stem cell (CSC) properties in TNBC. METHODS: Expression of the M2-like macrophage marker CD163 was evaluated by immunohistochemistry in human breast cancer tissues. The phenotype of M2 macrophages polarized from Tohoku-Hospital-Pediatrics-1 (THP1) cells was verified by flow cytometry. Transwell assays, wound healing assays, western blotting, flow cytometry, ELISA, quantitative polymerase chain reaction (qPCR), luciferase reporter gene assays, and immunofluorescence assays were conducted to investigate the mechanism by which TAMs regulate EMT and CSC properties in BT549 and HCC1937 cells. RESULTS: Clinically, we observed a high infiltration of M2-like tumor-associated macrophages in TNBC tissues and confirmed that TAMs were associated with unfavorable prognosis in TNBC patients. Moreover, we found that conditioned medium from M2 macrophages (M2-CM) markedly promoted EMT and CSC properties in BT549 and HCC1937 cells. Mechanistically, we demonstrated that chemokine (C-C motif) ligand 2 (CCL2) secretion by TAMs activated Akt signaling, which in turn increased the expression and nuclear localization of ß-catenin. Furthermore, ß-catenin knockdown reversed TAM-induced EMT and CSC properties. CONCLUSIONS: This study provides a novel mechanism by which TAMs promote EMT and enhance CSC properties in TNBC via activation of CCL2/AKT/ß-catenin signaling, which may offer new strategies for the diagnosis and treatment of TNBC. Video Abstract.


Assuntos
Quimiocina CCL2 , Transição Epitelial-Mesenquimal , Células-Tronco Neoplásicas , Proteínas Proto-Oncogênicas c-akt , Neoplasias de Mama Triplo Negativas , Macrófagos Associados a Tumor , beta Catenina , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Quimiocina CCL2/metabolismo , Humanos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Microambiente Tumoral , Macrófagos Associados a Tumor/metabolismo , Macrófagos Associados a Tumor/patologia , beta Catenina/metabolismo
3.
Oncogenesis ; 11(1): 19, 2022 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-35459265

RESUMO

Previously, our lab explored that tongue cancer resistance-associated protein (TCRP1) plays a central role in cancer chemo-resistance and progression. Absolutely, TCRP1 was significantly increased in lung cancer. But the mechanism is far from elucidated. Here, we found that TCRP1 was increased in p53-mutant non-small-cell lung cancer (NSCLC), comparing to that in NSCLC with wild type p53. Further study showed that mutant p53 couldn't bind to the promoter of TCRP1 to inhibit its expression. While the wild type p53 did so. Next, loss-and gain-of-function assays demonstrated that TCRP1 promoted cell proliferation and tumor growth in NSCLC. Regarding the mechanism, TCRP1 encouraged AKT phosphorylation and blocked FOXO3a nuclear localization through favoring FOXO3a ubiquitination in cytoplasm, thus, promoted cell cycle progression. Conclusionly, TCRP1 was upregulated in NSCLC cells with mutant p53. TCRP1 promoted NSCLC progression via regulating cell cycle.

4.
Front Oncol ; 11: 607989, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33889541

RESUMO

Glioblastoma (GBM), the primary malignant brain tumor, is typically associated with a poor prognosis and poor quality of life, mainly due to the lack of early diagnostic biomarkers and therapeutic targets. However, gene sequencing technologies and bioinformatics analysis are currently being actively utilized to explore potential targets for the diagnosis and management of malignancy. Herein, based on a variety of bioinformatics tools for the reverse prediction of target genes associated with the prognosis of GBM, a ceRNA network of AGAP2-AS1-miR-9-5p-MMP2/MMP9 was constructed, and a potential therapeutic target for GBM was identified. Enrichment analysis predicted that the ceRNA regulatory network participates in the processes of cell proliferation, differentiation, and migration.

5.
Front Genet ; 12: 617350, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33767729

RESUMO

Glioblastoma accounts for 45.2% of central nervous system tumors. Despite the availability of multiple treatments (e.g., surgery, radiotherapy, chemotherapy, biological therapy, immunotherapy, and electric field therapy), glioblastoma has a poor prognosis, with a 5-year survival rate of approximately 5%. The pathogenesis and prognostic markers of this cancer are currently unclear. To this end, this study aimed to explore the pathogenesis of glioblastoma and identify potential prognostic markers. We used data from the GEO and TCGA databases and identified five genes (ITGA5, MMP9, PTPRN, PTX3, and STX1A) that could affect the survival rate of glioblastoma patients and that were differentially expressed between glioblastoma patients and non-tumors groups. Based on a variety of bioinformatics tools for reverse prediction of target genes associated with the prognosis of GBM, a ceRNA network of messenger RNA (STX1A, PTX3, MMP9)-microRNA (miR-9-5p)-long non-coding RNA (CRNDE) was constructed. Finally, we identified five potential therapeutic drugs (bacitracin, hecogenin, clemizole, chrysin, and gibberellic acid) that may be effective treatments for glioblastoma.

6.
Cell Death Differ ; 27(3): 966-983, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31296961

RESUMO

Breast cancer stem cells (BCSCs) are tumor initiating cells that can self-renew and are highly tumorigenic and chemoresistant. Therefore, the identification of factors critical for BCSC function is vital for the development of therapies. Here, we report that DNMT1-mediated FOXO3a promoter hypermethylation leads to downregulation of FOXO3a expression in breast cancer. FOXO3a is functionally related to the inhibition of FOXM1/SOX2 signaling and to the consequent suppression of BCSCs properties and tumorigenicity. Moreover, we found that SOX2 directly transactivates DNMT1 expression and thereby alters the methylation landscape, which in turn feedback inhibits FOXO3a expression. Inhibition of DNMT activity suppressed tumor growth via regulation of FOXO3a/FOXM1/SOX2 signaling in breast cancer. Clinically, we observed a significant inverse correlation between FOXO3a and FOXM1/SOX2/DNMT1 expression levels, and loss of FOXO3a expression or increased expression of FOXM1, SOX2, and DNMT1 predicted poor prognosis in breast cancer. Collectively, our findings suggest an important role of the DNMT1/FOXO3a/FOXM1/SOX2 pathway in regulating BCSCs properties, suggesting potential therapeutic targets for breast cancer.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinogênese/genética , Carcinogênese/patologia , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Regulação para Baixo , Proteína Forkhead Box O3/metabolismo , Células-Tronco Neoplásicas/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Metilação de DNA/genética , Regulação para Baixo/genética , Retroalimentação Fisiológica , Feminino , Proteína Forkhead Box M1/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos Nus , Células-Tronco Neoplásicas/patologia , Regiões Promotoras Genéticas , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais
7.
J Cancer ; 10(24): 5964-5974, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31762806

RESUMO

Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer subtype and lacks effective targeted therapies. Cancerous inhibitor of protein phosphatase 2A (Cip2a) is an oncogene that is known to inhibit PP2A tumor suppressor activity in human malignancies. We previously demonstrated that Cip2a is a novel target for the treatment of TNBC. However, the functional roles of Cip2a in TNBC progression are still not fully characterized. In this study, we identified that miR-301a is a novel target of Cip2a in TNBC cell lines by miRNA microarray analysis. We found that Cip2a increases E2F1 expression, which in turn transcriptional activates miR-301a by occupying the miR-301a host gene SKA2 promoter. Moreover, we found that miR-301a level is significantly increased in TNBC tissues, and up-regulation of miR-301a is responsible for Cip2a-induced cell proliferation and invasion of TNBC cells. Furthermore, miR-301a feedback promotes the expression of Cip2a via activation of ERK/CREB signaling. Together, our study suggests an auto-regulatory feedback loop between Cip2a and miR-301a and this auto-regulatory loop might play an important role in TNBC progression.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA