Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Biochim Pol ; 69(4): 855-864, 2022 Dec 12.
Article in English | MEDLINE | ID: mdl-36508480

ABSTRACT

This study aimed to demonstrate that chlorogenic acid (CGA) has anticancer effects against ovarian cancer. The MTT assay was used to assess the optimum concentrations of CGA on the ovarian cancer cell lines OVCA433 and SKOV3, followed by the rate of apoptosis using Annexin V-FITC/PI. The mitochondrial membrane potential of ovarian tumour cells treated with CGA was evaluated using mitochondrial staining kits followed by Western blot analysis, immunofluorescence, and RT-PCR assays. The Trans-well migration assay conducted the percentage of cell migration, followed by wound healing and colony formation assays. CGA induces activation of mitochondria-mediated intrinsic apoptotic pathways in ovarian cancer cells. The discovery that miR-199a-5p is inversely correlated to DDR1, a receptor tyrosine kinase involved in collagen synthesis, was the major consequence of examining the various mechanisms involved in the development of ovarian cancer. After treatment with CGA, cells derived from ovarian cancer cells were deregulated partially via the miR199a5p/DDR1 axis, significantly affecting tumour suppression. DDR1 has been identified as a direct target of miR199a5p in these ovarian cancer cells. We found that CGA-induced loss of DDR1 caused the inactivation of NF-κB signalling downstream in the MMP, migration, and EMT pathways. The study results showed that CGA is a promising drug candidate for treating ovarian cancer, particularly because it exhibits anti-invasive and migrastatic properties.


Subject(s)
MicroRNAs , Ovarian Neoplasms , Humans , Female , Chlorogenic Acid/pharmacology , Discoidin Domain Receptor 1/metabolism , Cell Movement , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Apoptosis , Cell Line, Tumor , Cell Proliferation , MicroRNAs/metabolism
2.
Acta Biochim Pol ; 69(1): 245-250, 2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35231164

ABSTRACT

Pimaric acid is a naturally occurring resin and has been found to perform many pharmacological activities including, anticancer activity. However, the role of Pimaric acid in ovarian cancer is still not known. This investigation aimed to evaluate the anticancer effects of Pimaric acid and its molecular mechanism in human ovarian cancer cells. MTT assay was used to examine cell viability. Cell morphology was determined through phase contrast microscopy. DAPI staining and TUNEL assay were performed for apoptotic study. Examination of cell cycle phase distribution was carried out through flow cytometry. In vitro wound healing assay was used for cell migration determination. Pimaric acid induced cytotoxicity in human ovarian cancer cells (PA-1) in a dose-dependent manner without causing too much cytotoxicity in human ovarian epithelial cells (T1074). Cell morphology in treated cancer cells showed significant changes compared to untreated controls. Furthermore, it was observed that the cytotoxic effects of Pimaric acid were apoptosis-mediated and caspase-dependent cascade. Western blotting analysis showed that the expression of apoptosis-associated proteins like BAX, p-53 and caspase-3 was enhanced and BCL-2 expression was diminished. The induction of cytotoxicity was mediated via endoplasmic reticulum stress through expressions of related proteins which showed a tremendous increase in p-PERK, PERK, AT-4, CHOP and IRE-1 levels after treatment. Cell cycle analysis through cytometry showed significant results as it revealed G2/M phase cell cycle arrest. Furthermore, the in vitro wound healing assay showed specific anti-migratory effects of Pimaric acid on PA-1 cells. In conclusion it can be assumed that Pimaric acid may act as a potential anticancer agent against ovarian carcinoma, however further investigations are required to validate this initial claim.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Diterpenes/pharmacology , Endoplasmic Reticulum Stress/drug effects , Ovarian Neoplasms/pathology , Caspases/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism
3.
Med Sci Monit ; 27: e932350, 2021 Mar 25.
Article in English | MEDLINE | ID: mdl-33762566

ABSTRACT

Retracted, due to breach of publishing guidelines, following the identification of non-original and manipulated figure images. Reference: Wang Li, Zhou Ping, Gao Xuemei, Luo Minglian, Meng Hongjuan, He Yi, Zhu Zhongxiang: Naturally Occurring Sclareol Diterpene Augments the Chemosensitivity of Human Hela Cervical Cancer Cells by Inducing Mitochondrial Mediated Programmed Cell Death, S-Phase Cell Cycle Arrest and Targeting Mitogen-Activated Protein Kinase (MAPK)/Extracellular-Signal-Regulated Kinase (ERK) Signaling Pathway. Med Sci Monit 2020; 26:e920248. 10.12659/MSM.920248.

4.
Med Sci Monit ; 26: e920248, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31935210

ABSTRACT

BACKGROUND Cervical cancer is a major threat to female health worldwide. This study was performed to study the anticancer potential of sclareol and as a chemo-sensitizing agent against human cervical cancer cells along with evaluating its effects on apoptosis, cell cycle arrest, and MAPK/ERK signaling pathway. MATERIAL AND METHODS MTT assay was performed to check cell viability, morphological changes were observed through phase-contrast microscopy, DAPI (4',6-diamidino-2-phenylindole) staining and TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labelling) assays were performed to evaluate apoptotic effects; MMP (matrix metalloproteinase) and cell cycle analysis were examined through flow cytometry. Western blotting analysis was performed to check the protein expressions of MAPK/ERK signaling pathway and apoptosis proteins. RESULTS Results depicted that both sclareol and cisplatin induced cytotoxic effects individually but when used in combination, it led to much more pronounced cytotoxic effects indicating a synergistic effect of sclareol on cisplatin. Sclareol treatment led to significant decrease in the levels of p-MEK and p-ERK. Significant morphological changes (including chromatin condensation, nuclear fragmentation) in cervical cancer cells were seen after treatment. Western blot showed significant alterations including increase in BAX and decrease in BCL-2 levels. An increase in the S-phase cells, indicating cell cycle arrest at S-phase was seen along with modulating the expressions of CDK-1and Cdc25C, and increase in the levels of p-CDK-1, cyclin-B1, cyclin-A, and p-Cdc25C. CONCLUSIONS Sclareol not only induced cytotoxic effects but also enhanced chemosensitivity of human cervical cancer cells towards cisplatin and these effects are mediated via MAPK/ERK signaling pathway, stimulation of apoptosis and S-phase cell cycle arrest.


Subject(s)
Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Diterpenes/pharmacology , MAP Kinase Signaling System/drug effects , Mitochondria/metabolism , S Phase/drug effects , Uterine Cervical Neoplasms/enzymology , Uterine Cervical Neoplasms/pathology , Cell Cycle Proteins/metabolism , Cell Shape/drug effects , Diterpenes/chemistry , Female , HeLa Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...