Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Anticancer Agents Med Chem ; 23(19): 2102-2110, 2023.
Article in English | MEDLINE | ID: mdl-37723632

ABSTRACT

BACKGROUND: Gastric cancer is one of the most common and deadliest malignancies in the world. Therefore, there is an urgent need to develop new and effective agents to reduce mortality. The plants of genus Inula have gained the attention of researchers worldwide as a rich source of potent medicinal compounds. OBJECTIVE: This study explores the anti-cancer activity of Britannin, a sesquiterpene lactone isolated from Inula aucheriana DC., and its molecular mechanism in gastric cancer cells, AGS and MKN45. METHODS: Cytotoxicity was evaluated through the MTT assay following 24 h, 48 h, and 72 h treatment with different concentrations of Britannin. Apoptosis rate and caspase-3 activity were measured 24 h after treatment by Britannin. . Western blotting was performed to determine the expression of the NF-κB, IκBα, and PPARγ proteins. Moreover, quantitative RT-PCR was applied to measure the expression of NF-κB target genes. RESULTS: We showed that Britannin induced cell growth inhibition and apoptosis in gastric cancer cells. Britannin caused an elevation in mRNA and protein levels of PPARγ. The involvement of PPARγ was more confirmed using GW9662, a PPARγ inhibitor. Suppression of NF-κB was demonstrated by western blot analysis. Down-regulation of MMP-9, TWIST-1, COX-2, and Bcl-2 and up-regulation of Bax were also observed in gastric cancer cells. CONCLUSION: These results imply that activation of the PPARγ signaling pathway through suppression of NF-κB underlies the anti-cancer properties of Britannin in gastric cancer. Therefore, Britannin could be considered as a promising anti-cancer candidate for further evaluation.


Subject(s)
Inula , Sesquiterpenes , Stomach Neoplasms , Humans , NF-kappa B/metabolism , PPAR gamma/genetics , Stomach Neoplasms/drug therapy , Signal Transduction , Lactones/pharmacology , Sesquiterpenes/pharmacology , Apoptosis , Cell Line, Tumor
2.
J Ethnopharmacol ; 281: 114529, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34418511

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The genus Inula has been traditionally used as folk medicine in treating different illnesses such as kidney stones, urethra infection, jaundice, bronchitis, respiratory diseases and cancer. AIM OF THE STUDY: Gaillardin, a sesquiterpene lactone isolated from Inula oculus-christi, seems to have great potential as an anti-cancer agent. This study was carried out to evaluate the anti-cancer properties of Gaillardin in gastric cancer cells and also its possible underlying mechanism. METHODS: The colorimetric MTT assay was used to assess metabolic activity of cells as an indicator of viability and cytotoxicity. Flow cytometry using Annexin V-FITC/PI was applied to detect and quantify the level of apoptosis. Detection of activated caspase-3, as a biochemical marker of apoptosis, was done using caspase-3 assay kit. Activation of NF-κB pathway was determined by western blotting. The mRNA expression levels of NF-κB target genes were measured using quantitative RT-PCR. Moreover, intracellular reactive oxygen species (ROS) production was evaluated. RESULTS: Gaillardin significantly reduced cell viability in a time and dose-dependent manner. The inhibitory effect of Gaillardin was attributed to induction of apoptosis. Investigation about the underlying mechanism revealed that Gaillardin exerts its action through inhibition of NF-κB activation and subsequently down-regulation of genes (COX-2, MMP-9, TWIST-1, and BCl-2) regulated by NF-κB. Moreover, Gaillardin caused remarkable elevation in ROS production in AGS and MKN45 cells. CONCLUSION: We provided evidences about the role of NF-κB pathway in the induction of apoptosis by the sesquiterpene lactone Gaillardin in AGS and MKN45 cells for the first time, which suggest Gaillardin could be considered as a promising natural agent for further investigations to identify new potent anticancer drugs in the future.


Subject(s)
Apoptosis/drug effects , Down-Regulation/drug effects , Gene Expression Regulation/drug effects , Lactones/pharmacology , NF-kappa B/metabolism , Sesquiterpenes/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Humans , Inula/chemistry , Lactones/chemistry , Molecular Structure , NF-kappa B/genetics , Plant Components, Aerial/chemistry , Sesquiterpenes/chemistry
3.
Nutr Cancer ; 73(10): 2030-2038, 2021.
Article in English | MEDLINE | ID: mdl-32933316

ABSTRACT

Ovarian cancer due to the difficulties in early clinical diagnosis and absence of successfull treatment has poor prognosis and high mortality among all gynecological malignancies. Many evidence has revealed that plants of the Euphorbiaceae family are precious sources of novel bioactive compounds with anti-tumor activities. 3,7,14,15-tetraacetyl-5-propanoyl-13(17)-epoxy-8,10(18)-myrsinadiene (TPEM) is a new myrsinane-type diterpene isolated recently by our group from aerial parts of Euphorbia Connata and the present study was aimed to explore its inhibitory effects on growth of two human ovarian cancer cells, OVCAR-3, and Caov-4. The obtained results indicated that growth of OVCAR-3 and Caov-4 cells was significantly inhibited by TPEM in a dose-dependent manner, with the IC50 values of 41.27 ± 1.52 and 36.44 ± 2.41 µM, respectively. Furthermore, using Annexin V-FITC and PI staining it was confirmed that the induced cell death was mainly mediated through apoptotic pathway. Further observations such as decrease in the mitochondrial membrane potential (ΔΨm), increase in the activity of caspase-3 and elevation of the Bax/Bcl-2 ratio suggested the role of mitochondria in the induction of apoptosis by TPEM. ROS level was also remarkably increased in OVCAR-3 and Caov-4 cells in response to TPEM treatment. In conclusion, these findings provide first evidences about potential anticancer properties of TPEM.


Subject(s)
Diterpenes , Euphorbia , Ovarian Neoplasms , Apoptosis , Cell Line, Tumor , Diterpenes/pharmacology , Humans , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Reactive Oxygen Species/metabolism
4.
Onco Targets Ther ; 11: 7669-7677, 2018.
Article in English | MEDLINE | ID: mdl-30464515

ABSTRACT

BACKGROUND: Drimia maritima (D. maritima) is a plant belonging to the family Asparagaceae, which has been used for the treatment of several ailments including cancer around the world. To our knowledge, there is no comprehensive study about the molecular mechanisms of anticancer activity of this plant, yet. MATERIALS AND METHODS: In the current study, cell viability, apoptosis induction, ROS production, mitochondrial apoptotic pathway, and ER stress mediators have been evaluated in breast cancer cells, MCF7, and MDA-MB-468 treated with D. maritima. RESULTS: Significant cytotoxic effects were observed in MCF-7 and MDA-MB-468 cells after exposure to D. maritima. Apoptosis induction was determined using Annexin-V-FITC and propidium iodide staining. Furthermore, an increase of ROS, loss of mitochondrial membrane potential, the release of cytochrome c, activation of caspases, and elevation in the Bax/Bcl-2 ratio was determined. D. maritima dose-dependently increased the mRNA expression of ER stress markers such as CHOP, ATF-4, GADD34, and TRIB3 in MCF-7, and MDA-MB-468 cells. CONCLUSION: These data suggest that D. maritima induces apoptosis in human breast cancer cells via the mitochondrial-mediated pathway. In addition, endoplasmic reticulum stress seems to be involved in D. maritima-induced cell death.

5.
Artif Cells Nanomed Biotechnol ; 45(8): 1769-1777, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28278581

ABSTRACT

Novel formulations of nanocomposites derived from ZnO nanoparticles have provided potential biomedical applications as a new strategy for treatment of breast cancer. In this research, two types of ZnO nanomaterials were synthesized by sol-gel hydrothermal process and co-precipitation containing fast quenching and also surface modification methods. The cytotoxic effects on growth of the breast cancer cell lines MCF-7 were evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. Cell viability of the breast cancer cell line MCF-7 was reduced with increasing ZnO nanofluid concentrations at 48 and 72 h of treatment. The IC50 value of MCF-7 cells after 72 h of treatment with the first product ZnO (a) and second one ZnO


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Iron/chemistry , Nanocomposites/chemistry , Silver/chemistry , Water/chemistry , Zinc Oxide/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Compounding , Humans , MCF-7 Cells , Nanoparticles/chemistry
6.
Cell Biol Toxicol ; 31(6): 295-305, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26843455

ABSTRACT

Medicinal plant extracts have been widely used for cancer treatment. Gaillardin is a natural sesquiterpene lactone that has recently been reported to have anticancer properties. The ability to induce apoptosis is an important property of a candidate anticancer drug, which discriminates between anticancer drugs and toxic compounds. The current study was therefore carried out to address the issue if Gaillardin is able to induce apoptosis in the breast cancer cell lines MCF-7 and MDA-MB-468 and to determine the underlying mechanism of its anticancer effects. Apoptosis induction by Gaillardin treatment was confirmed by annexin V-FITC/PI staining, and caspase-3,-6, and-9 activation. Using Western blot analysis, we found that Gaillardin upregulated the pro-apoptotic protein Bax and p53 and downregulated the anti-apoptotic protein Bcl-2. Moreover, the apoptotic effect of Gaillardin was also related to ROS production and loss of mitochondrial membrane potential (ΔΨm). Taken together, these results demonstrate that Gaillardin can inhibit proliferation of breast cancer cells via inducing mitochondrial apoptotic pathway and therefore, might be a promising molecule in cancer chemoprevention or chemotherapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Inula/chemistry , Lactones/pharmacology , Sesquiterpenes/pharmacology , Antineoplastic Agents, Phytogenic/isolation & purification , Caspase 3/genetics , Caspase 3/metabolism , Caspase 6/genetics , Caspase 6/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Iran , Lactones/isolation & purification , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Plants, Medicinal , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Sesquiterpenes/isolation & purification , Signal Transduction , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
7.
Tumour Biol ; 36(2): 1191-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25342596

ABSTRACT

Induction of apoptosis in cancer cells can be a promising treatment method in cancer therapy. Naturally derived products had drawn growing attention as agent in cancer therapy. The main target of anticancer drugs may be distinct, but eventually, they lead to identical cell death pathway, which is apoptosis. Here, we indicated that britannin, a sesquiterpene lactone isolated from Asteraceae family, has antiproliferative activity on the MCF-7 and MDA-MB-468 human breast cancer cells. Annexin V/propidium iodide (PI) staining, Hoechst 33258 staining, and caspase-3/9 activity assay confirmed that britannin is able to induce apoptosis in MCF-7 and MDA-MB-468 cells. The Western blot analysis showed that the expression of Bcl-2 was noticeably decreased in response to britannin treatment, while the expression of Bax protein was increased, which were positively correlated with elevated expression of p53. Moreover, britannin also increased reactive oxygen species (ROS) generation which in turn triggered the loss of mitochondrial transmembrane potential (ΔΨm) and the subsequent release of cytochrome c from mitochondria into cytosol. Taken together, these results suggest that britannin inhibits growth of MCF-7 and MDA-MB-468 breast cancer cells through the activation of the mitochondrial apoptotic pathway and may potentially serve as an agent for breast cancer therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Lactones/administration & dosage , Sesquiterpenes/administration & dosage , Breast Neoplasms/pathology , Caspase 3/biosynthesis , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , bcl-2-Associated X Protein/biosynthesis
8.
Cell Biol Int ; 38(2): 228-38, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24155029

ABSTRACT

The inhibitory effects of acetazolamide on the growth and proliferation of epithelial breast cancer cells (T-47D) were investigated. Analysis of morphological changes indicated little apoptosis in T-47D cells incubated with acetazolamide, according to data from flow cytometry, DNA laddering, and expression of AIF. However, an increase in caspase-3 activity was detected in cells. This was concomitant with an increase in DFF45/DFF40 ratio leading to inhibition of caspase-3 activity, DNA fragmentation and progression of apoptosis. Flow cytometry also confirmed that acetazolamide had no significant effect on cell cycle progression. These results are consistent with lack of change in the expression of cell cycle regulatory proteins p21, p27, cdc2 and cyclinD1. Increased expression of ATG5, p53 and DRAM, along with an increase in BCLN1/Bcl-2 ratio, indicated that acetazolamide inhibited the proliferation of T-47D cells by inducing autophagy. Increased expression of PTEN, along with decreased expression of Akt1, also showed that acetazolamide treatment resulted in death inducing autophagy. Collectively the results indicate that autophagy is an adequate mechanism mediating the anti-cancer effects of acetazolamide in T-47D cells through engagement of p53/DRAM pathway and attenuation of Akt survival signalling.


Subject(s)
Acetazolamide/pharmacology , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Breast Neoplasms/drug therapy , Breast/drug effects , Carbonic Anhydrase Inhibitors/pharmacology , Anticonvulsants/pharmacology , Apoptosis/drug effects , Breast/metabolism , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , DNA Fragmentation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...