Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 13(1): 10845, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37407632

ABSTRACT

The average survival of patients with glioblastoma is 12-15 months. Therefore, finding a new treatment method is important, especially in cases that show resistance to treatment. Extremely low-frequency electromagnetic fields (ELF-EMF) have characteristics and capabilities that can be proposed as a new cancer treatment method with low side effects. This research examines the antitumor effect of ELF-EMF on U87 and U251 glioblastoma cell lines. Flowcytometry determined the viability/apoptosis and distribution of cells in different phases of the cell cycle. The size of cells was assessed by TEM. Important cell cycle regulation genes mRNA expression levels were investigated by real-time PCR. ELF-EMF induced apoptosis in U87cells much more than U251 (15% against 2.43%) and increased G2/M cell population in U87 (2.56%, p value < 0.05), and S phase in U251 (2.4%) (data are normalized to their sham exposure). The size of U87 cells increased significantly after ELF-EMF exposure (overexpressing P53 in U251 cells increased the apoptosis induction by ELF-EMF). The expression level of P53, P21, and MDM2 increased and CCNB1 decreased in U87. Among the studied genes, MCM6 expression decreased in U251. Increasing expression of P53, P21 and decreasing CCNB1, induction of cell G2/M cycle arrest, and consequently increase in the cell size can be suggested as one of the main mechanisms of apoptosis induction by ELF-EMF; furthermore, our results demonstrate the possible footprint of P53 in the apoptosis induction by ELF-EMF, as U87 carry the wild type of P53 and U251 has the mutated form of this gene.


Subject(s)
Glioblastoma , Humans , Glioblastoma/genetics , Glioblastoma/pathology , Tumor Suppressor Protein p53/genetics , Electromagnetic Fields/adverse effects , M Cells , G2 Phase Cell Cycle Checkpoints/genetics
2.
Prog Biophys Mol Biol ; 182: 49-58, 2023 09.
Article in English | MEDLINE | ID: mdl-37302516

ABSTRACT

Recent studies have shown that non-ionizing electromagnetic fields (NIEMFs) in a specific frequency, intensity, and exposure time can have anti-cancer effects on various cancer cells; however, the underlying precise mechanism of action is not transparent. Most cancer deaths are due to metastasis. This important phenomenon plays an inevitable role in different steps of cancer including progression and development. It has different stages including invasion, intravasation, migration, extravasation, and homing. Epithelial-mesenchymal transition (EMT), as well as hybrid E/M state, are biological processes, that involve both natural embryogenesis and tissue regeneration, and abnormal conditions including organ fibrosis or metastasis. In this context, some evidence reveals possible footprints of the important EMT-related pathways which may be affected in different EMFs treatments. In this article, critical EMT molecules and/or pathways which can be potentially affected by EMFs (e.g., VEGFR, ROS, P53, PI3K/AKT, MAPK, Cyclin B1, and NF-кB) are discussed to shed light on the mechanism of EMFs anti-cancer effect.


Subject(s)
Epithelial-Mesenchymal Transition , Signal Transduction , Cell Line, Tumor , Phosphatidylinositol 3-Kinases , Electromagnetic Fields , Cell Movement
3.
Eur J Pharmacol ; 910: 174454, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34454929

ABSTRACT

Thyroid cancer is the most common type of endocrine-related cancer. According to the literature, its incidence is not very high, but its rate increasing especially in developed countries. With this regard, finding approaches to prevent, and exert anti-tumor activity with the least side effects on the normal cells at the next step after diagnosis is demanded. Herbal medicine is a branch of integrative oncology that seems to be a practically beneficial goddess for cancer treatment in many cases. Here we utilized Hypericin (HYP) to investigate its anti-tumor (apoptotic and anti-metastatic) activity on B-CPAP (a thyroid cancer cell line) and cytotoxicity on TPC-1 (thyroid cancer cell line with wild type TP53) cell lines. To assess whether HYP may exert preventive and anti-tumor effects and does not have a potential side effect, we dubbed the experiments on the fibroblast cells (as a normal cell line). Cytotoxicity and kind of cellular death were examined by MTT and AnnexinV/PI respectively. Extrinsic/intrinsic apoptosis pathway induction was clarified by western blotting on pro/cleaved caspases 9, 8, and 3. According to our data HYP induces an extrinsic apoptosis pathway and no other types (necroptosis, necrosis, etc.) in B-CPAP cells. Moreover, CDH1 mRNA expression calculated to be up-regulated, and that of LGALS3 down-regulated in the B-CPAP cell line after treatment. Besides tumor cytotoxic activity, we suggest that HYP impedes with invasion and/or metastasis process.


Subject(s)
Anthracenes/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Perylene/analogs & derivatives , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/metabolism , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Inhibitory Concentration 50 , Neoplasm Metastasis , Perylene/pharmacology , Signal Transduction/drug effects , Thyroid Neoplasms/pathology
4.
Life Sci ; 266: 118874, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33358904

ABSTRACT

AIMS: Hypericin (HYP) from Hypericum perforatum has cytotoxic effects on a variety of malignant cell types, but the pattern of gene expression mediating the effect is largely unknown. Here we sought to analyze the response of U87 glioblastoma (GBM) cell lines in response to HYP. MATERIALS AND METHODS: U87 cell line was treated by HYP. Cytotoxicity was assessed using MTT and Annexin V/PI assays. Gene expression profile was obtained using high-throughput sequencing. Enrichment analysis was performed on differentially expressed genes (DEGs). Upstream transcription factors and microRNAs regulating DEGs were predicted. The effects of DEGs on survival of GBM patients were calculated. Protein-protein interaction analysis was conducted to obtain key altered genes. The possible effect of HYP treatment on immunity response was evaluated. KEY FINDINGS: The IC50 of HYP on U87 cell line was determined to be 1.5 µg/ml. The main type of cell death was apoptosis. A total of 312 DEGs were found. Affected Gene Ontology terms and pathways were identified. Analysis of upstream modulators of DEGs pointed out to transcription factors that significantly overlap with GBM stem cell transcription factor. Survival analysis suggested that HYP works best for the mesenchymal subtype patients. Tumor infiltration analysis predicted that HYP may affect Treg and macrophage infiltration in vivo. Using expression pattern of GBM patients and HYP-induced DEGs we suggested Fedratinib as a complementary drug to HYP. SIGNIFICANCE: Our study represents the response of U87 cell line to HYP, with analyses on survival, transcription factors and personalization according to GBM subtype.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/drug therapy , Perylene/analogs & derivatives , Transcriptome/drug effects , Anthracenes , Apoptosis , Cell Proliferation , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Perylene/pharmacology , Prognosis , Survival Rate , Tumor Cells, Cultured
5.
Exp Eye Res ; 190: 107883, 2020 01.
Article in English | MEDLINE | ID: mdl-31758976

ABSTRACT

Retinal pigment epithelial (RPE) cells are indispensable for eye organogenesis and vision. To realize the therapeutic potential of in vitro-generated RPE cells for cell-replacement therapy of RPE-related retinopathies, molecular mechanisms of RPE specification and maturation need to be investigated. So far, many attempts have been made to decipher the regulatory networks involved in the differentiation of human pluripotent stem cells into RPE cells. Here, we exploited a highly-efficient RPE differentiation protocol to determine global expression patterns of microRNAs (miRNAs) during human embryonic stem cell (hESC) differentiation into RPE using small RNA sequencing. Our results revealed a significant downregulation of pluripotency-associated miRNAs along with a significant upregulation of RPE-associated miRNAs in differentiating cells. Our functional analyses indicated that two RPE-enriched miRNAs (i.e. miR-125b and let-7a) could promote RPE fate at the expense of neural fate during RPE differentiation. Taken together, these mechanistic interrogations might shed light on a better understanding of RPE cell development and provide insights for the future application of these cells in regenerative medicine.


Subject(s)
Cell Differentiation/physiology , MicroRNAs/genetics , Retinal Pigment Epithelium/cytology , Cell Line , Flow Cytometry , Gene Expression Profiling , Human Embryonic Stem Cells/metabolism , Humans , Immunohistochemistry , MicroRNAs/physiology , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Phagocytosis/physiology , Real-Time Polymerase Chain Reaction , Retinal Pigment Epithelium/metabolism , Signal Transduction
6.
Arch Iran Med ; 21(9): 387-392, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30221528

ABSTRACT

BACKGROUND: Breast cancer is the major cause of death from cancer among women around the world. Given the drug resistance in the treatment of this disease, it is very important to identify new therapies and anticancer drugs. Many studies demonstrated that hypericin could induce apoptosis in different cancer cell lines; however, the underlying mechanism is not well understood yet. Therefore, this study aimed to evaluate the anticancer effect of hypericin in two breast cancer cell lines, one with wild type P53 and the other with mutant P53. METHODS: In this study, the MDA-MB-231 and MDA-MB-175-VII cell lines were treated with different concentrations of hypericin for 24 and 48 hours. The measurement of cell death was performed by MTT assay. The cell apoptosis rate was measured using annexin V/propidium iodide assay through flow cytometry. The level of expression in P21 and P53 genes was evaluated by real time PCR. Immunocytochemistry (ICC) analysis was performed for P21 (direct target for P53 protein) to confirm the results. RESULTS: The results showed that hypericin could have dose-dependent cytotoxic effects on the MDA-MB-231 and MDA-MB-175-VII cell lines, and its cytotoxicity is much higher in the latter cells. According to flow cytometry results, 86% of MDA-MB-175-VII cells underwent apoptosis with IC50 dose of hypericin for MDA-MB-231 cells after 24 hours. Moreover, after 24 hours of exposure to hypericin with MDA- MB-231 IC50 concentration, the expression of P53 and P21 genes upregulated in MDA-MB-175-VII much more than MDA-MB-231 when both cell lines were treated with 24 hours IC50 dose of MDA-MB-231. The ICC analysis on P21 confirmed that by treating both cell lines with MDA-MB-231 IC50 dose of hypericin for 24 hours, this protein is overexpressed much more in MDA-MB-175-VII cells. CONCLUSION: The results of this study demonstrated that hypericin's apoptotic and cytotoxic effects on cancer cells may be mediated via P53 overexpression, cell cycle arrest and the subsequent apoptosis. Therefore, it is of great importance to consider that hypericin would have better impact on cells or tumors with wild type P53.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/pathology , Perylene/analogs & derivatives , Tumor Suppressor Protein p53/genetics , Anthracenes , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Perylene/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...