Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Recent Pat Anticancer Drug Discov ; 17(4): 416-426, 2022.
Article in English | MEDLINE | ID: mdl-35049439

ABSTRACT

BACKGROUND: The incidence of melanoma has been increasing over the last 30 years. The most common treatments, such as surgery, chemotherapy, and radiotherapy, frequently cause serious damage to the body. It is therefore critical to develop a new therapeutic strategy for the treatment of melanoma. OBJECTIVES: This research aims to evaluate the anti-tumor effect of Neochamaejasmine A (NCA) on B16F10 melanoma cells and the underlying molecular mechanisms. METHODS: The CCK-8 kit was utilized to assay the influence of NCA on the vitality of B16F10 cells. Modifications in B16F10 cells morphology were observed using a phase-contrast microscope. Apoptosis of B16F10 melanoma cells was assessed by Hoechst 33258, Annexin V and propidium iodide staining. Cell cycle was detected using a commercial kit by flow cytometry. The mRNA and protein expression levels associated with apoptosis and cell cycle arrest were detected by RT-PCR and Western blot. The expression level of pathway proteins was assessed using Western blot. RESULTS: It was found that the proliferation of B16F10 cells was inhibited by NCA in concentration- and time-dependent manners. NCA promoted apoptosis by halting the cell cycle at the G2/M phase. After treatment with NCA, cell apoptosis was confirmed by Hoechst 33258 staining. NCA triggered the cell cycle to seize at the G2/M stage by downregulating cyclin B1 and cyclin-dependent kinase 2 (CDC2) expression. Moreover, the mRNA and protein expression of cleaved caspase- 9 and Bcl-2-associated X-protein (Bax) were increased, whereas there was a decline in the expression of B-cell lymphoma 2 (Bcl-2). The p-p38/p38 and phosphorylated c-Jun N-terminal kinase (p-JNK/JNK) ratio were also elevated by NCA. The apoptosis and G2/M cell cycle arrest were inhibited in cells co-treated with the p38 inhibitor SB203580 and JNK inhibitor SP600125. The expression of apoptosis-related proteins Bax was decreased, and Bcl-2 was increased. CONCLUSION: The findings of this study showed that NCA could induce apoptosis and cell cycle arrest in B16F10 melanoma cells by activating JNK and p38 MAPK signaling pathway.


Subject(s)
Apoptosis , Cell Cycle , Medicine, Chinese Traditional , Melanoma , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , G2 Phase Cell Cycle Checkpoints , Humans , Melanoma/pathology , Melanoma/therapy , Mitogen-Activated Protein Kinases , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Oxid Med Cell Longev ; 2020: 3237150, 2020.
Article in English | MEDLINE | ID: mdl-32411325

ABSTRACT

The botanical constituents of Stellera chamaejasme Linn. exhibit various pharmacological and medicinal activities. Neochamaejasmin A (NCA), one main active constituent of S. chamaejasme, inhibits cell proliferation and induces cell apoptosis in several types of tumor cells. However, the antitumor effect of NCA on hepatocellular carcinoma cells is still unclear. In this study, NCA (36.9, 73.7, and 147.5 µM) significantly inhibited hepatoblastoma-derived HepG2 cell proliferation in a concentration-dependent manner. Hoechst 33258 staining and flow cytometry showed that apoptotic morphological changes were observed and the apoptotic rate was significantly increased in NCA-treated HepG2 cells, respectively. Additionally, the levels of Bax, cleaved caspase-3, and cytoplasmic cytochrome c were increased, while the level of Bcl-2 was decreased in NCA-treated HepG2 cells when compared with the control group. Moreover, we found that the reactive oxygen species (ROS) level was significantly higher and the mitochondrial membrane potential was remarkably lower in NCA-treated HepG2 cells than in the control group. Further studies demonstrated that the levels of p-JNK and p-ERK1/2 were significantly upregulated in NCA-treated HepG2 cells, and pretreatment with JNK and ERK1/2 inhibitors, SP600125 and PD0325901, respectively, suppressed NCA-induced cell apoptosis of HepG2 cells. In addition, NCA also significantly inhibited human hepatoma BEL-7402 cell proliferation and induced cell apoptosis through the ROS-mediated mitochondrial apoptotic pathway. These results implied that NCA induced mitochondrial-mediated cell apoptosis via ROS-dependent activation of the ERK1/2/JNK signaling pathway in HepG2 cells.


Subject(s)
Apoptosis/drug effects , Biflavonoids/pharmacology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , MAP Kinase Signaling System , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Antioxidants/metabolism , Biflavonoids/chemistry , Carcinoma, Hepatocellular/enzymology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Shape/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Liver Neoplasms/enzymology , MAP Kinase Signaling System/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects
3.
Front Oncol ; 10: 415, 2020.
Article in English | MEDLINE | ID: mdl-32300554

ABSTRACT

Chamaejasmin B (CHB), a natural biflavone isolated from Stellera chamaejasme L., has been reported to exhibit anti-cancer properties; however, its effect in melanoma cells is not clear. Here, we aimed to investigate the anticancer effect of CHB in mouse melanoma B16F0 and B16F10 cells. We found that CHB significantly suppressed cell proliferation and promoted cell cycle arrest at G0/G1 phase in B16F0 cells; it also induced cell differentiation and increased melanin content by increasing tyrosinase (TYR) activity and mRNA levels of melanogenesis-related genes in B16F0 cells. Meanwhile, wound closure, invasion, and migration of B16F0 and B16F10 cells were dramatically inhibited. Moreover, CHB significantly increased ROS levels and decreased ΔΨm, resulting in B16F0 and B16F10 cell apoptosis. Finally, in vivo studies showed that CHB inhibited tumor growth and induced tumor apoptosis in a mouse xenograft model of murine melanoma B16F0 and B16F10 cells. Overall, CHB decreases malignant characteristics and may be a promising therapeutic agent for malignant melanoma cells via multiple signaling pathways.

4.
Front Pharmacol ; 10: 285, 2019.
Article in English | MEDLINE | ID: mdl-31001113

ABSTRACT

The first-line chemotherapy drug adriamycin (ADM) is widely used for the treatment of breast cancer, but the acquired drug resistance and the normal tissue toxicity remain clinical challenges. Alteronol has been reported to exert wide-ranging anti-tumor activity. In this study, we firstly examined the synergistic anti-tumor effects and the underlying mechanisms of alteronol combined with ADM in breast cancer. We have found that the combination of alteronol and ADM significantly suppressed the expression levels of the cell cycle-related proteins (CDC2 and Cyclin B1) and induced cell cycle arrest at the G2/M phase, leading to cell proliferation inhibition in breast cancer 4T1 cells. Moreover, co-treatment of alteronol and ADM (i) remarkably activated p38 and JNK kinases, (ii) elevated ROS levels, (iii) triggered mitochondrial dysfunction, (iv) released cytochrome c into the cytoplasm, (v) upregulated apoptosis-related proteins, e.g., cleaved PARP, Bax, and cleaved caspase-3/9, and (vi) downregulated the expression of Bcl-2, followed by apoptosis. Furthermore, our in vivo studies showed that the low-dose combination of alteronol (2 mg/kg) and ADM (1 mg/kg) significantly inhibited tumor growth in tumor bearing mice, and the anti-tumor effect of the combination was the same as that of high-dose ADM (8 mg/kg). In addition, the low-dose combination group showed lower toxicities to major organs than the high-dose ADM group. Taken together, these data demonstrate that the low-dose combination of alteronol and ADM could notably improve the anti-tumor activity and have lower toxicities to major organs than those in high-dose ADM group.

5.
J Pharm Pharmacol ; 70(4): 516-524, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29411391

ABSTRACT

OBJECTIVE: Emerging evidence showed that alteronol has a potential antitumour effect in several tumour cells. However, the antitumour effect of alteronol on breast cancer has not been reported. This study investigated the mechanisms of alteronol-induced cell proliferation inhibition in human breast cancer T47D cells. METHODS: After treatment with alteronol, T47D cell proliferation was examined by MTT assay. The cell cycle distribution, cell apoptosis, reactive oxygen species level and mitochondrial membrane potential were evaluated via flow cytometry. Next, the protein levels of cyclin B1, cdc2, p21, p-cyclin B1, p-cdc2, p53, Bax, Bcl-2 and cytochrome c were analysed using Western blot analysis. Meanwhile, the mRNA levels of cyclin B1, cdc2, p21 and p53 were examined by qRT-PCR. KEY FINDINGS: Our data showed that alteronol inhibited the proliferation of T47D cells via inducing G2-phase arrest and cell apoptosis. Compared with control group, alteronol significantly increased ROS level and triggered mitochondrial dysfunction in alteronol-treated T47D cells. Further studies showed that the mRNA and protein levels of cdc2 and cyclin B1 were downregulated, while the mRNA and protein levels of p21, p53, p-cyclin B1, p-cdc2 and cytochrome c were upregulated. In addition, the expression level of Bax was increased, and the expression level of Bcl-2 was decreased. CONCLUSIONS: Alteronol induced T47D cell cycle arrest and cell apoptosis through increasing ROS production and triggering mitochondrial dysfunction, and subsequently inhibiting T47D cell proliferation.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/metabolism , Cell Cycle Checkpoints/drug effects , Naphthoquinones/pharmacology , Reactive Oxygen Species/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/physiology , Breast Neoplasms/drug therapy , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Dose-Response Relationship, Drug , Female , Humans , Naphthoquinones/therapeutic use , Reactive Oxygen Species/agonists
SELECTION OF CITATIONS
SEARCH DETAIL
...