Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Appl Toxicol ; 2024 Jul 14.
Article in English | MEDLINE | ID: mdl-39004823

ABSTRACT

The aim of this study was to evaluate the anticancer effects of piceatannol, a natural stilbenoid, on human neuroblastoma cells. In order to accomplish this goal, we performed various cellular assays, including the XTT cell proliferation assay for cell viability, colony formation assay for colony formation capacity, FITC Annexin V and cell death detection kit for apoptosis, matrigel invasion assay for invasion capacity, intracellular reactive oxygen species (ROS) red dye for intracellular ROS levels, TMRM staining method for mitochondrial membrane potential (MMP), and the CYTO-ID autophagy detection kit for autophagy. Furthermore, we analyzed the expression levels of genes associated with apoptosis and autophagy using RT-qPCR. Based on our findings, piceatannol exhibited cytotoxic effects on neuroblastoma cells. Besides, treatment with piceatannol at both 50 and 100 µM concentrations for 72 h decreased colony formation, induced apoptosis and autophagy, inhibited cell invasion, decreased MMP, and increased ROS levels in SH-SY5Y cells. In addition, we observed significant upregulation in the expression levels of CASP8, BECLIN, ATG5, ATG7, and MAPILC3A genes between the two doses. These results suggest that piceatannol enhances autophagic activity and induces caspase-dependent apoptosis, indicating its potential as a therapeutic agent against neuroblastoma cells.

2.
World J Microbiol Biotechnol ; 40(7): 204, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38755413

ABSTRACT

Globally colorectal cancer ranks as the third most widespread disease and the third leading cause of cancer-associated mortality. Immunotherapy treatments like PD-L1 blockade have been used to inhibit the PD-L1 legend, which boosts the activity of cytotoxic T lymphocytes. Recently, studies suggest that some probiotics could potentially enhance the effectiveness of immunotherapy treatments for cancer patients. We found that in Caco-2 and HT-29 cells, the live Leuconostoc mesenteroides treatment resulted an increase in the PD-L1 expression and this treatment stimulated interferon-gamma (IFN-γ) production in Jurkat T-cells. Due to the well-established ability of IFN-γ to enhance PD-L1 expression, the combination of IFN-γ and L. mesenteroides was used in colon cancer cell lines and a resulting remarkable increase of over tenfold in PD-L1 expression was obtained. Interestingly, when L. mesenteroides and IFN-γ are present, the blockage of PD-L1 using PD-L1 antibodies not only improved the viability of Jurkat T-cells but also significantly boosted the levels of IFN-γ and IL-2, the T-cells activation marker cytokines. In addition to upregulating PD-L1, L. mesenteroides also activated Toll-like receptors (TLRs) and NOD-like receptors (NODs) pathways, specifically through TLR2 and NOD2, while also exerting a suppressive effect on autophagy in colon cancer cell lines. In conclusion, our findings demonstrate a significant upregulation of PD-L1 expression in colon cancer cells upon co-culturing with L. mesenteroides. Moreover, the presence of PD-L1 antibodies during co-culturing activates Jurkat T cells. The observed enhancement in PD-L1 expression may be attributed to the inhibition of the Autophagy pathway or activation of the hippo pathway. KEY POINTS: Co-culturing L. mesenteroides increases PD-L1 gene and protein transaction in colon cancer. L. mesenteroides existing enhances T cells viability and activity. GPCR41/42 is a possible link between L. mesenteroides, YAP-1 and PD-L1.


Subject(s)
B7-H1 Antigen , Colonic Neoplasms , Interferon-gamma , Leuconostoc mesenteroides , Up-Regulation , Humans , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Interferon-gamma/metabolism , Colonic Neoplasms/immunology , HT29 Cells , Jurkat Cells , Caco-2 Cells , Leuconostoc mesenteroides/metabolism , Leuconostoc mesenteroides/genetics , Interleukin-2/metabolism , Lymphocyte Activation , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Probiotics/pharmacology , Cell Line, Tumor , Nod2 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/genetics
3.
Mol Biol Rep ; 51(1): 340, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38393422

ABSTRACT

BACKGROUND: Treatment of Pancreatic Cancer (PC) is challenging due to its aggressiveness and acquired resistance to conventional chemotherapy and radiotherapy. Therefore, the discovery of new therapeutic agents and strategies is essential. Juglone, a naphthoquinone, is a secondary metabolite produced naturally in walnut-type trees having allelopathic features in its native environment. Juglone was shown to prevent cell proliferation and induce ROS-mediated mitochondrial apoptosis. Ascorbate with both antioxidant and oxidant features, shows selective cytotoxicity in cancer cells. METHODS AND RESULTS: In this study, we evaluated the anticancer effects of Juglone in combination with ascorbate in PANC-1 and BxPC-3 PC cells. The MTT assay was used to determine the IC50 dose of Juglone with 1 mM NaAscorbate (Jug-NaAsc). Subsequently, the cells were treated with 5, 10, 15 and 20 µM Jug-NaAsc for 24 h. Apoptotic effects were evaluated by analyzing the following genes using qPCR; proapoptotic Bax, antiapoptotic Bcl-2 related to the mitochondrial apoptotic pathway and apoptosis inhibitor Birc5 (Survivin). Immunofluorescence analysis was performed using Annexin V-FITC in PC cells. As an antioxidant enzyme, Trx2 protein levels were determined by a commercial ELISA test kit. Jug-NaAsc treatment decreased the expressions of antiapoptotic genes Bcl-2 and Birc5 while the apoptotic gene Bax expression increased at all doses. Additionally, a dose-dependently increase of apoptosis according to immunofluorescence analysis and the decreases of Trx2 enzyme levels at all treatments in both cell lines supported gene expression results. CONCLUSION: Our results suggest that Juglone is a potential anticancer agent especially when combined with ascorbate.


Subject(s)
Naphthoquinones , Pancreatic Neoplasms , Humans , Reactive Oxygen Species/metabolism , Antioxidants/pharmacology , Antioxidants/therapeutic use , bcl-2-Associated X Protein/metabolism , Cell Line, Tumor , Apoptosis , Ascorbic Acid/pharmacology , Naphthoquinones/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics
4.
Med Oncol ; 40(11): 315, 2023 Oct 03.
Article in English | MEDLINE | ID: mdl-37787808

ABSTRACT

Neuroblastoma is the most common brain solid tumor in infancy. Despite the availability of numerous approaches like immunotherapy, surgery, chemotherapy, and radiotherapy, neuroblastoma frequently develops resistance and recurs. Immunotherapy is one of the most promising approaches and PD-L1 antibody blocking is the phenomena used to inhibit PD-1 receptors to increase and improve cytotoxic T cells toward cancer. Numerous studies underlined the critical role of probiotics on immune system development and modulation in addition to possible role in inducing apoptosis in cancer cells. In this study, a Streptococcus thermophilus strain, isolated from a local yogurt, was used as it is considered a potential probiotic due to its tolerance lower pH, bile acid, antibiotic suitability, and blood hemolysis. Our results showed that S. thermophilus lysates played as an immune checkpoint modulator at 25 µg/ml dose boosting PD-L1 transcripts and protein levels in SH-SY5Y neuroblastoma cell line. Interestingly, co-culture between SH-SY5Y and Jurkat T cells in the presence of blocking PD-L1 antibodies increased Jurkat T-cell viability compering to control without lysate. On the other hand, annexin-V/7-AAD, qPCR and western blot results showed that S. thermophilus lysates at 200 and 400 µg/ml decreased SH-SY5Y cell viability and increased apoptotic marker genes transcription and caspase-3 and caspase-9 protein expression.


Subject(s)
Brain Neoplasms , Neuroblastoma , Humans , B7-H1 Antigen , Streptococcus thermophilus , Neuroblastoma/therapy , Neoplasm Recurrence, Local , Apoptosis
5.
Mol Biol Rep ; 49(12): 11947-11957, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36260179

ABSTRACT

BACKGROUND: Piceatannol is a naturally occurring plant-derived phenolic compound (stilbenoid), an analogue of resveratrol. It has been shown that, piceatannol has biological activity properties such as antiproliferative, antioxidative, anti-inflammatory and proapoptotic, in various human cancer studies in vitro and in vivo. OBJECTIVES AND METHODS: In this study, it was aimed to investigate whether piceatannol induces apoptosis through anticancer activity methods (cell viability, colony formation, annexin-V/7-AAD, ROS (Reactive oxygen species), MMP (Mitochondrial membrane potential), wound healing, invasion assay, RT-qPCR (Real-Time Quantitative Polymerase Chain Reaction), western blotting in PANC-1 and MIA PaCa-2 pancreatic cancer (PC) cell lines. RESULTS: According to our results, piceatannol decreased cell viability in a dose and time-dependent manner [the half-maximal inhibitory concentration (IC50): 60 µM in PANC-1 and IC50: 90 µM in MIA PaCa-2 cell line at 48 h (h)] and caused significant changes in the expression of apoptosis-related genes and protein. Piceatannol induced apoptosis in PANC-1 and MIA PaCa-2 cells, accompanied by increased ROS production, decreased MMP, and increased Caspase-3-9 activity. Piceatannol also inhibited colony-forming abilities, invasion, and migration of PC cells. CONCLUSION: Our results show that piceatannol has an anti-cancerogenic effect on PANC-1 and MIA PaCa-2 cells, and exerts this effect by suppressing proliferation and inducing apoptosis. Therefore, piceatannol could be considered to be a potential chemotherapeutic agent candidate for the treatment and prevention of PC.


Subject(s)
Pancreatic Neoplasms , Stilbenes , Humans , Caspases/metabolism , Reactive Oxygen Species/metabolism , Up-Regulation , Cell Line, Tumor , Apoptosis , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Stilbenes/pharmacology , Cell Survival , Mitochondria/metabolism , Pancreatic Neoplasms
6.
Mol Neurobiol ; 59(11): 7036-7044, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36074231

ABSTRACT

Depression is one of the most important and serious health problems in developing countries which affects millions of people. It is associated with the decrease of the quality of life as well as suicides and mortality. The disease may show recurrent episodes in some patients. Obviously, not all the patients with depression could be treated properly, because some individuals are drug-resistant and the options for the therapy are limited. Therefore, it is crucial to investigate new molecules and pathways that may have possible antidepressant activity. Sirtuin (SIRT), known as a class III histone deacetylase, which is regulated by nicotinamide adenine dinucleotide (NAD +), is one of these molecules. In the current study, we investigated the possible antidepressant-like effect of SIRT2 inhibitor AK-7. For this purpose, behavioral tests were performed in chronic AK-7-treated mice, and the expression levels of BDNF, NGF, NTF3, CREB, NTRK2, ERK1, ERK2, and GAP43 genes were evaluated by qRT-PCR analysis in brain tissues. Protein levels for BDNF, CREB1, and NTRK2 were determined by western blot. Our data showed that AK-7 significantly decreased immobility time and showed antidepressant-like effect. In addition, AK-7 treatment significantly increased mRNA levels of CREB and NTRK2 and protein levels of CREB1, BDNF, and NTRK2. Finally, our results suggest that SIRT2 and AK-7 may have a potential role in the cellular mechanisms of depression.


Subject(s)
Sirtuin 2 , Suicide , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Brain-Derived Neurotrophic Factor/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Depression/drug therapy , Depression/genetics , Depression/metabolism , Humans , Mice , NAD/metabolism , Nerve Growth Factor/metabolism , Quality of Life , RNA, Messenger/metabolism , Sirtuin 2/metabolism , Up-Regulation/genetics
7.
Toxicol In Vitro ; 84: 105458, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35988885

ABSTRACT

The aim of this study was to determine possible anticancer effect of tomentosin, a natural sesquiterpene lactone, on pancreatic cancer cells. The cytotoxic effect of tomentosin was determined by XTT analysis. Colony formation and apoptosis analyzes were performed, Reactive oxygen species (ROS) level and change in mitochondrial membrane potential (MMP) were evaluated in control and tomentosin-treated cells. The effect of tomentosin on expression levels of apoptosis-related genes was determined by qRT-PCR and Caspase-3 and Caspase-9 proteins were analyzed by western blot. And, the effect of tomentosin on migration and invasion of cells were evaluated. The IC50 dose of tomentosin was found to be 31.11 µM in PANC-1 cells and 33.93 µM in MIA PaCa-2 cells for 48 h. And, treatment of tomentosin at IC50 dose suppressed the colony forming capacity of cells. While tomentosin increased apoptosis rate and ROS production, an decrease was observed in MMP. Tomentosin affected expression level of apoptosis-related genes and increased Caspase-3 and Caspase-9 protein levels. After tomentosin treatment, cell migration and invasion were suppressed. As a result, this study reveals that tomentosin has anticancer effects on pancreatic cancer cells, and therefore it predicts that tomentosin can be evaluated as an effective agent against pancreatic cancer.


Subject(s)
Pancreatic Neoplasms , Sesquiterpenes , Apoptosis , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Line, Tumor , Humans , Lactones/pharmacology , Membrane Potential, Mitochondrial , Pancreatic Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Sesquiterpenes/pharmacology , Pancreatic Neoplasms
8.
Gene ; 805: 145904, 2021 Dec 30.
Article in English | MEDLINE | ID: mdl-34418470

ABSTRACT

Breast cancer is the second most common cause of cancer-related mortality in women. Breast cancer metastasis which usually is observed at the last stage is the major cause of breast cancer-related death. Long non-coding RNAs (lncRNAs) are member of the non-coding RNA family. It is known that lncRNAs have important functions in the genes regulation of different processes and pathways such as EMT (Epithelial mesenchymal transition), metastasis and apoptosis. Therefore, it is inevitable that lncRNAs have potential contribution for the understanding of cancer pathogenesis. lncRNA-ZEB2NAT is the natural antisense transcript of ZEB2. Herein, we investigated the effects of lncRNA-ZEB2NAT on process of EMT, metastasis and apoptosis in MCF7 and MDA-MB-231 breast cancer cells. The effect of ZEB2NAT on the expression of important genes in EMT, metastasis and apoptosis, and some protein levels was determined by qRT-PCR and western blot analysis, respectively. The effects of ZEB2NAT on cell proliferation, apoptosis, invasion and colony formation were evaluated using XTT, annexin V, invasion and colony assays, respectively. The ZEB2NAT knockdown caused anti-metastatic and apoptotic effects. The ZEB2NAT knockdown resulted in a decrease in ZEB2 and N-cadherin but an increase in E-cadherin protein levels. In addition, it was determined that ZEB2NAT knockdown significantly decreased cell proliferation and stimulated apoptosis in both cells. It was found that ZEB2NAT knockdown significantly decreased invasion and colony formation in both cells. ZEB2NAT knockdown showed anti-metastatic and apoptotic effect by affecting the important genes in both cells. These results have suggested that ZEB2NAT has an important role in EMT, metastasis and apoptosis in breast cancer and ZEB2NAT knockdown caused significant anti-cancer activities.


Subject(s)
Breast Neoplasms/genetics , Zinc Finger E-box Binding Homeobox 2/genetics , Apoptosis/genetics , Breast Neoplasms/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , RNA, Long Noncoding/genetics , Zinc Finger E-box Binding Homeobox 2/metabolism
9.
Med Oncol ; 38(9): 113, 2021 Aug 11.
Article in English | MEDLINE | ID: mdl-34378101

ABSTRACT

The aim of this study was to determine the effect of lncRNA HIF1A-AS2 on autophagy-associated drug resistance in small cell lung cancer (SCLC) cells. The expression of HIF1A-AS2 was silenced by siRNA in doxorubicin-sensitive H69 and doxorubicin-resistant H69AR cells. Then, cytotoxicity, apoptosis and autophagy analyses were carried out in the normoxic and CoCl2-induced hypoxic environment. The effect of HIF1A-AS2 on the expression levels of genes, which are associated with drug resistance and autophagy, was determinated by qRT-PCR analysis. The levels of MRP1, HIF-1α and Beclin-1 were analyzed by western blot method. Knockdown of HIF1A-AS2 increased doxorubicin sensitivity of SCLC cells and decreased autophagy. Knockdown of HIF1A-AS2 has also affected the expression of several genes that will increase drug sensitivity and inhibit autophagy in both cell lines. The levels of HIF-1α and Beclin-1 were decreased in both cell lines by knockdown of HIF1A-AS2. MRP1 expression was decrease in H69AR cells. In addition, CoCl2-induced hypoxic environment decreased in doxorubicin sensitivity of H69 cells, and knockdown of HIF1A-AS2 reversed this effect of hypoxia. Knockdown of HIF1A-AS2 increased drug sensitivity of SCLC cells in relation to autophagy. Therefore, hypoxia-HIF1A-AS2-autophagy interaction is thought to be determinative in drug sensitivity of these cells.


Subject(s)
Autophagy , Biomarkers, Tumor/genetics , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , RNA, Long Noncoding/antagonists & inhibitors , Small Cell Lung Carcinoma/drug therapy , Antibiotics, Antineoplastic/pharmacology , Apoptosis , Cell Proliferation , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...