Cyanidin 3-O-Glucoside Induces the Apoptosis in the Osteosarcoma Cells through Upregulation of the PPARγ and P21: An In Vitro Study

2020 ◽  
Vol 20 (9) ◽  
pp. 1087-1093
Author(s):  
Hesam A. Atashi ◽  
Hamid Z. Arani ◽  
Amirhossein Shekarriz ◽  
Hamidreza Nazari ◽  
Amirhossein Zabolian ◽  
...  

Background: Osteosarcoma (OS) is known as the malignant tumors in the bone. Cyanidin 3-OGlucoside (C3G) has a potential to induce the apoptotic cell death in different cancer cells; however, the mechanisms of action for C3G have not been clarified yet. Objective: In this study, the apoptotic effects of C3G on three different osteosarcoma cell lines including Saso-2, MG-63, and G-292 (clone A141B1) were investigated. Methods: The 24-hr IC50 of C3G for Saso-2, G-292, and MG-63 cells was evaluated by the MTT assay. Apoptosis induction in these cell lines after treatment with the C3G was approved by the Annexin V/PI flow cytometry. Changes at the mRNA expression level of PPARγ, P21, Bax, and Bcl-xl genes were investigated by real-time Polymerase Chain Reaction (PCR) technique, and P21 expression was further confirmed by the western blotting. Results: The MTT assay results demonstrated that the 24-hr IC50 of C3G was equal to 110μg/ml for Saso-2 and G-292 cells while it was about 140μg/ml for the MG-63 cells. The results of real-time PCR clearly showed that treatment of the cells with 24hrs IC50 of C3G caused the upregulation of PPARγ, P21, and Bax genes. Moreover, western blot analysis confirmed that P21 protein overexpressed endogenously after treatment of the cells with the C3G, and it was more upregulated in the MG-63 cells compared to the other cell lines. Conclusion: According to the findings of the study, the C3G is a novel anti-osteosarcoma agent with the ability to induce the apoptosis in different osteosarcoma cells through upregulation of the PPARγ and P21 genes.

2020 ◽  
Vol 15 (1) ◽  
pp. 871-883
Author(s):  
Jinshan Zhang ◽  
Dan Rao ◽  
Haibo Ma ◽  
Defeng Kong ◽  
Xiaoming Xu ◽  
...  

AbstractBackgroundOsteosarcoma is a common primary malignant bone cancer. Long noncoding RNA small nucleolar RNA host gene 15 (SNHG15) has been reported to play an oncogenic role in many cancers. Nevertheless, the role of SNHG15 in the doxorubicin (DXR) resistance of osteosarcoma cells has not been fully addressed.MethodsCell Counting Kit-8 assay was conducted to measure the half-maximal inhibitory concentration value of DXR in osteosarcoma cells. Western blotting was carried out to examine the levels of autophagy-related proteins and GDNF family receptor alpha-1 (GFRA1). Quantitative reverse transcription-polymerase chain reaction was performed to determine the levels of SNHG15, miR-381-3p, and GFRA1. The proliferation of osteosarcoma cells was measured by MTT assay. The binding sites between miR-381-3p and SNHG15 or GFRA1 were predicted by Starbase bioinformatics software, and the interaction was confirmed by dual-luciferase reporter assay. Murine xenograft model was established to validate the function of SNHG15 in vivo.ResultsAutophagy inhibitor 3-methyladenine sensitized DXR-resistant osteosarcoma cell lines to DXR. SNHG15 was upregulated in DXR-resistant osteosarcoma tissues and cell lines. SNHG15 knockdown inhibited the proliferation, DXR resistance, and autophagy of osteosarcoma cells. MiR-381-3p was a direct target of SNHG15, and GFRA1 bound to miR-381-3p in osteosarcoma cells. SNHG15 contributed to DXR resistance through the miR-381-3p/GFRA1 axis in vitro. SNHG15 depletion contributed to the inhibitory effect of DXR on osteosarcoma tumor growth through the miR-381-3p/GFRA1 axis in vivo.ConclusionsSNHG15 enhanced the DXR resistance of osteosarcoma cells through elevating the autophagy via targeting the miR-381-3p/GFRA1 axis. Restoration of miR-381-3p expression might be an underlying therapeutic strategy to overcome the DXR resistance of osteosarcoma.


NANO ◽  
2020 ◽  
Vol 15 (12) ◽  
pp. 2050163
Author(s):  
Hongkun Gao ◽  
Ping Fan ◽  
Qizhen Xu ◽  
Yiting Li ◽  
Jianxin Wang ◽  
...  

Melanoma, one of the most malignant tumors, is difficult to treat due to its high drug resistance. Silver nanoparticles (AgNPs) are widely used as antimicrobial agents in biomedical fields. In this study, the spherical AgNPs with average sizes of 5[Formula: see text]nm were prepared using a dopamine reduction method. The in vitro study shows that AgNPs with the concentrations of 0.5[Formula: see text][Formula: see text]g/mL and 1[Formula: see text][Formula: see text]g/mL exhibit good biocompatibility to 3T3L1 fibroblast cells. AgNPs with the same concentrations significantly inhibited the growth of B16 melanoma cells. In culture with B16 cells, AgNPs induced intracellular oxidative stress by generating the reactive oxygen species and reducing the superoxide dismutase, which further reduces the mitochondrial membrane potential. Moreover, the damage in mitochondria could activate mitochondrion-mediated cell apoptosis. The B16 cells apoptosis was analyzed by FITC-Annexin V/propidium iodide double staining assay, which confirms that AgNPs caused the abundance of apoptotic cells in different stages. Thus, AgNPs displayed the antitumor activity in vitro. Then, the therapeutic efficacy in vivo was evaluated in mice-bearing B16 melanoma tumors. The obtained results show the antitumor ability of AgNPs and provide a potential strategy for cancer treatment.


2008 ◽  
Vol 253 (1-2) ◽  
pp. 71-80 ◽  
Author(s):  
Michela Muraro ◽  
Oana M. Mereuta ◽  
Francesco Saglio ◽  
Francesca Carraro ◽  
Massimo Berger ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2393-2393
Author(s):  
Edgar Jost ◽  
Claudia Schubert ◽  
Tim Bruemmendorf ◽  
Oliver Galm

Abstract Abstract 2393 Poster Board II-370 Introduction: Hypermethylation of CpG islands in the promoter region of genes is a well characterized epigenetic modification associated with transcriptional silencing of cancer related genes and plays a crucial role in carcinogenesis. In addition, acetylation of core histones is necessary for the maintenance of transcriptional activity of genes. DNA methylation and histone deacetylation are reversible and can be influenced by DNA methyltransferase (DNMT) inhibitors such as 5-aza-2`-deoxycytidine (DAC) or 5-azacytidine (AZA) and histone deacetylase (HDAC) inhibitors such as suberoylanilide hydroxamic acid (SAHA), respectively. Clinical trials using a strategy based on the modification of epigenetic changes with DAC or AZA in combination with HDAC inhibitors have been promising and may help to generate new strategies in treatment of hematopoietic malignancies including multiple myeloma (MM). In MM however, only limited data are published about the possible synergistic effects between DNMT inhibitors and the highly potent pan-HDAC inhibitor SAHA. Material and Methods: To assess the in vitro effects of SAHA on the MM cell lines U266, LP-1, RPMI8226 or OPM-2 and the possible interactions with DNMT inhibitors, cells were first incubated with DAC in a final concentration of 0.1 or 0.2 mM for 72 hours. After exposure to DAC, cells were incubated for 72 or 96 hours with SAHA in a final concentration between 0.1 and 20 mM. The toxic effect of the treatment was assessed by an 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The drug concentration inducing a 50 % killing of the cells compared to control cell survival was calculated from the dose-response curve (IC50). Induction of apoptosis was analysed by flow cytometry with annexin V-binding. In addition, the expression of the epigenetically silenced tumor suppressor genes SFRP-2 and DAB2 was determined by real time RT-PCR before and after exposure to DAC and SAHA. Results: In LP-1 and U-266 cells, no relevant enhancement in the cytotoxic effect of SAHA was observed after previous exposure to DAC. In contrast, in OPM-2 and RPMI-8226 cells, a significant increase in cytotoxicity of SAHA was observed, when the cells were first incubated with DAC with a decrease of the IC50 from 6.5 μM to 2.43 μM and 10.37 μM to 4.5 μM, respectively. We further analysed a possible synergism between SAHA and DAC for the induction of apoptosis by flow cytometry. After sequential exposure of the cells with DAC for 72 hours and with SAHA for 72 hours, no change in the apoptotic cell fraction was observed for the cell lines OPM-2 and RPMI-8226. However, for U-266 and LP-1, a significant increase in apoptotic cells was observed after incubation with SAHA, when the cells were previously exposed to DAC with a increase in the apoptotic cell fraction of 39.5 % to 55.4 % and 2.5 % to 14.4 %, respectively. By real-time RT-PCR, corresponding transcriptional silencing for SFRP-2 and DAB2 was demonstrated in untreated cells, and exposure of cell lines to DAC and SAHA resulted in reexpression. A synergism for the induction of reexpression of these genes was observed when cells were incubated with DAC and SAHA sequentially. Discussion: After treatment with SAHA, we observed a dose-dependent induction of cell death and apoptosis as assessed by MTT and annexin V assay, respectively. In the different MM cell lines, we observed a synergism between SAHA and DAC both for cytotoxic effects and the induction of apoptosis. A synergism was also observed for the reexpression of epigenetically silenced genes after exposure to DAC and SAHA. These in vitro data can be considered as a basis for further in vitro studies and preclinical models with SAHA in combination with demethylating agents such as DAC in order to improve treatment response and survival in MM patients. Disclosures: Jost: MSD: Research Funding. Bruemmendorf:Genzyme: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees.


Toxins ◽  
2019 ◽  
Vol 11 (9) ◽  
pp. 506 ◽  
Author(s):  
Tony Haykal ◽  
Peter Nasr ◽  
Mohammad H. Hodroj ◽  
Robin I. Taleb ◽  
Rita Sarkis ◽  
...  

Annona cherimola Mill is a large green fruit with black seeds widely known to possess toxic properties due to the presence of Annonaceous acetogenins. The present study investigates the anti-cancer properties of an Annona cherimola Mill ethanolic seed extract on Acute Myeloid Leukemia (AML) cell lines in vitro and elucidates the underlying cellular mechanism. The anti-proliferative effects of the extract on various AML cell lines and normal mesenchymal cells (MSCs) were assessed using WST-1 viability reagent. The pro-apoptotic effect of the extract was evaluated using Annexin V/PI staining and Cell Death ELISA. The underlying mechanism was deciphered by analyzing the expression of various proteins using western blots. Treatment with an A. cherimola seed ethanolic extract promotes a dose- and time-dependent inhibition of the proliferation of various AML cell lines, but not MSCs. Positive Annexin V staining, as well as DNA fragmentation, confirm an increase in apoptotic cell death by upregulating the expression of pro-apoptotic proteins which control both intrinsic and extrinsic pathways of apoptosis. GC/MS analysis revealed the presence of phytosterols, in addition to other bioactive compounds. In conclusion, Annona cherimola Mill seed extract, previously known to possess a potent toxic activity, induces apoptosis in AML cell lines by the activation of both the extrinsic and the intrinsic pathways.


2017 ◽  
Vol 24 (3) ◽  
pp. 147-156 ◽  
Author(s):  
Kirk Jensen ◽  
Athanasios Bikas ◽  
Aneeta Patel ◽  
Yevgeniya Kushchayeva ◽  
John Costello ◽  
...  

The HIV protease inhibitor Nelfinavir (NFV) inhibits PI3K/AKT and MAPK/ERK signaling pathways, emerging targets in thyroid cancers. We examined the effects of NFV on cancer cells that derived from follicular (FTC), papillary (PTC) and anaplastic (ATC) thyroid cancers. NFV (1–20 µM) was tested in FTC133, BCPAP and SW1736 cell lines. The effects of NFV on cell proliferation were determined in vitro using real-time microscopy and by flow cytometry. DNA damage, apoptotic cell death and expression of molecular markers of epithelial–mesenchymal transition (EMT) were determined by Western blot and real-time PCR. Real-time imaging demonstrated that NFV (10 µM) increased the time required for the cell passage through the phases of cell cycle and induced DNA fragmentation. Growth inhibitory effects of NFV were associated with the accumulation of cells in G0/G1 phase, downregulation of cyclin D1 and cyclin-dependent kinase 4 (CDK4). NFV also induced the expression of γH2AX and p53BP1 indicating DNA damage. Treatment with NFV (20 µM) resulted in caspase-3 cleavage in all examined cells. NFV (20 µM) decreased the levels of total and p-AKT in PTEN-deficient FTC133 cells. NFV had no significant effects on total ERK and p-ERK in BRAF-positive BCPAP and SW1736 cells. NFV had no effects on the expression of EMT markers (Twist, Vimentin, E- and N-Cadherin), but inhibited the migration and decreased the abilities of thyroid cancer cells to survive in non-adherent conditions. We conclude that NFV inhibits proliferation and induces DNA damage in thyroid cancer cell lines. Our in vitro data suggest that NFV has a potential to become a new thyroid cancer therapeutic agent.


2017 ◽  
Vol 6 (04) ◽  
pp. 5360 ◽  
Author(s):  
Rajkiran Reddy Banala ◽  
Satish Kumar Vemuri ◽  
Gurava Reddy A.V. ◽  
Subbaiah G.P.V.*

Psoriasis is a chronic inflammatory skin disorder characterized by rapid proliferation of keratinocytes and incomplete keratinization. Discovery of safer and more effective anti-psoriatic drugs remains an area of active research at the present time. A431 and B16-F10 cell lines were used as in vitro models. In the present study, we aimed at assessing the Anti-psoriatic activity of aqueous extract of Acalypha indica. We analyzed the efficiency of A. indica leaf extract in inducing cell death and apoptosis in these cell lines. The cell death (Propidium iodide) and apoptosis (Annexin V) was assessed by fluorescence studies and we observed 80% of cell death and 75% of apoptosis in both cell lines. Therefore, this in vitro study suggested that the leaf extract is capable of serving as anti-psoriasis agent or compound.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3019-3019
Author(s):  
Lavinia Biamonte ◽  
Cinzia Federico ◽  
Eugenio Morelli ◽  
Emanuela Leone ◽  
Maria Eugenia Gallo Cantafio ◽  
...  

Abstract MicroRNAs (miRNAs), short non-coding RNAs which tune gene expression at post-transcriptional level, are recently emerging as key players in pathogenesis, progression and drug-resistance of multiple myeloma (MM). In this disease, they can act either with tumor-promoting or tumor-suppressing functions, depending on the nature of target mRNAs. Nowadays, effective strategies are available both to replace or to inhibit the expression of deregulated miRNAs, thus prompting the design of miRNA-based therapeutic strategies. We have recently demonstrated that miR-125b has tumor suppressor activity in MM and that enforced expression of synthetic miR-125b-5p mimics induces significant anti-MM activity in vitro and in vivo by targeting cell addiction to IRF4/cMyc pro-survival signaling. Moreover, we uncovered a functional feedback loop between cMyc and miR-125b in MM cells, whereas cMyc directly suppresses miR-125b transcription which, in turn, negatively regulates cMyc expression by targeting IRF4 mRNA. In the present study, we investigated the therapeutic potential of synthetic miR-125b-5p mimics combined with cMyc targeting agents, including the 10058-F4 small molecule inhibitor of cMyc-Max heterodimerization and the BET-bromodomain inhibitor JQ1, which is reported to inhibit cMyc transcription. At this aim, 3 MM cell lines (NCI-H929, SK-MM-1 and RPMI-8226) transfected with either miR-125b-5p mimics or scrambled oligonucleotides (miR-NC) were exposed to 10058-F4 (ranging from 10 to 100 μM) or JQ1 (ranging from 0,1 to 2μM) or DMSO. Effects on cell proliferation were then evaluated by CCK-8 assay at 24h, 48h and 72h time points, while the occurrence of apoptotic cell death was assessed by Annexin V flow-cytometry assay. Importantly, we found that enforced expression of miR-125b-5p mimics significantly and synergistically (synergistic index, SI >1) increases growth-inhibitory and pro-apoptotic activities of both 10058-F4 and JQ1. Similar results were observed in SK-MM-1 cells co-transfected with miR-125b-5p and cMyc siRNAs, while cMyc-defective U266 cells were not sensitized to either 10058-F4 nor JQ1 upon transfection with miR-125b-5p mimics. Furthermore, combinatorial treatments with JQ1 and miR-125b-5p mimics resulted in a stronger downregulation of cMyc protein, as compared to single molecules alone. Indeed, these results confirmed that impairment of cMyc activity/expression mediates the anti-MM synergistic effects between 10058-F4 or JQ1 and overexpression of miR-125b-5p by synthetic mimics. In conclusion, our data demonstrate a cMyc-mediated synergistic anti-MM activity of synthetic miR-125b-5p mimics with 10058-F4 or JQ1 cMyc targeting agents, providing the rationale for a more advanced preclinical investigations for the design of early clinical trials. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document