Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Life Sci ; 324: 121715, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37100377

ABSTRACT

AIMS: We aimed to evaluate the effect of periplocin on inhibiting hepatocellular carcinoma (HCC) and further determine its mechanisms. MAIN METHODS: Cytotoxic activity of periplocin against HCC cells was tested by CCK-8 and colony formation assays. The antitumor effects of periplocin were evaluated in human HCC SK-HEP-1 xenograft and murine HCC Hepa 1-6 allograft mouse models. Flow cytometry was used to measure cell cycle distribution, apopotosis, and the number of myeloid-derived suppressor cells (MDSCs). Hoechst 33258 dye was applied to observe the nuclear morphology. Network pharmacology was performed to predict possible signaling pathways. Drug affinity responsive target stability assay (DARTS) was used to evaluate AKT binding of periplocin. Western blotting, immunohistochemistry, and immunofluorescence were used to examine the protein expression levels. KEY FINDING: Periplocin inhibited cell viability with IC50 values from 50 nM to 300 nM in human HCC cells. Periplocin disrupted cell cycle distribution and promoted cell apoptosis. Moreover, AKT was predicted as the target of periplocin by network pharmacology, which was confirmed by that AKT/NF-κB signaling was inhibited in periplocin-treated HCC cells. Periplocin also inhibited the expression of CXCL1 and CXCL3, leading to decreased accumulation of MDSCs in HCC tumors. SIGNIFICANCE: These findings reveal the function of periplocin in inhibiting HCC progression by G2/M arrest, apoptosis and suppression of MDSCs accumulation through blockade of the AKT/NF-κB pathway. Our study further suggests that periplocin has the potential to be developed as an effective therapeutic agent for HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Myeloid-Derived Suppressor Cells , Humans , Mice , Animals , Carcinoma, Hepatocellular/pathology , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Liver Neoplasms/pathology , Myeloid-Derived Suppressor Cells/metabolism , Cell Proliferation , Apoptosis , Cell Line, Tumor
2.
Recent Pat Anticancer Drug Discov ; 17(3): 312-321, 2022.
Article in English | MEDLINE | ID: mdl-34951372

ABSTRACT

BACKGROUND: Cervical cancer is the fourth most prevalent gynecological cancer worldwide, which threatens women's health and causes cancer-related mortality. In the search for effective anticervical cancer drugs, we discovered that ß-estradiol (E2), a potent drug for estrogen deficiency syndrome treatment, displays the most potent cytotoxicity against HeLa cells. OBJECTIVE: This study aims to evaluate the growth inhibitory effect of ß-estradiol on HeLa cells and explore its underlying mechanisms. METHODS: CCK-8 assay was used to evaluate the cytotoxicity of 6 compounds against HeLa cells. Flow cytometric analysis and Hoechst 33258 staining assay were performed to detect cell cycle arrest and apoptosis induction. The collapse of the mitochondrial potential was observed by the JC-1 staining assay. The expression levels of proteins were examined by western blotting. RESULTS: ß-Estradiol, at high concentration, displays potent cytotoxicity against HeLa cells with an IC50 value of 18.71 ± 1.57 µM for 72 h treatment. ß-Estradiol induces G2/M cell cycle arrest through downregulating Cyclin B1 and p-CDK1. In addition, ß-estradiol-induced apoptosis is accompanied by the loss of mitochondrial potential, activation of the Caspase family, and altered Bax/Bcl-2 ratio. ß-Estradiol markedly decreased the expression level of p-AKT and p-NF-κB. CONCLUSION: This study demonstrated that ß-estradiol induces mitochondrial apoptosis in cervical cancer through the suppression of AKT/NF-κB signaling pathway, indicating that ß-estradiol may serve as a potential agent for cervical cancer treatment.


Subject(s)
NF-kappa B , Uterine Cervical Neoplasms , Apoptosis , Cell Line, Tumor , Cell Proliferation , Estradiol/pharmacology , Estradiol/therapeutic use , Female , HeLa Cells , Humans , NF-kappa B/metabolism , NF-kappa B/pharmacology , NF-kappa B/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Uterine Cervical Neoplasms/drug therapy
3.
Front Cell Dev Biol ; 9: 791882, 2021.
Article in English | MEDLINE | ID: mdl-34966744

ABSTRACT

Angiogenesis is required for tumor growth and development. Extracellular vesicles (EVs) are important signaling entities that mediate communication between diverse types of cells and regulate various cell biological processes, including angiogenesis. Recently, emerging evidence has suggested that tumor-derived EVs play essential roles in tumor progression by regulating angiogenesis. Thousands of molecules are carried by EVs, and the two major types of biomolecules, noncoding RNAs (ncRNAs) and proteins, are transported between cells and regulate physiological and pathological functions in recipient cells. Understanding the regulation of EVs and their cargoes in tumor angiogenesis has become increasingly important. In this review, we summarize the effects of tumor-derived EVs and their cargoes, especially ncRNAs and proteins, on tumor angiogenesis and their mechanisms, and we highlight the clinical implications of EVs in bodily fluids as biomarkers and as diagnostic, prognostic, and therapeutic targets in cancer patients.

SELECTION OF CITATIONS
SEARCH DETAIL
...