scholarly journals Profiling of O-acetylated Gangliosides Expressed in Neuroectoderm Derived Cells

2020 ◽  
Vol 21 (1) ◽  
pp. 370 ◽  
Author(s):  
Sumeyye Cavdarli ◽  
Nao Yamakawa ◽  
Charlotte Clarisse ◽  
Kazuhiro Aoki ◽  
Guillaume Brysbaert ◽  
...  

The expression and biological functions of oncofetal markers GD2 and GD3 were extensively studied in neuroectoderm-derived cancers in order to characterize their potential as therapeutic targets. Using immunological approaches, we previously identified GD3, GD2, and OAcGD2 expression in breast cancer (BC) cell lines. However, antibodies specific for O-acetylated gangliosides are not exempt of limitations, as they only provide information on the expression of a limited set of O-acetylated ganglioside species. Consequently, the aim of the present study was to use structural approaches in order to apprehend ganglioside diversity in melanoma, neuroblastoma, and breast cancer cells, focusing on O-acetylated species that are usually lost under alkaline conditions and require specific analytical procedures. We used purification and extraction methods that preserve the O-acetyl modification for the analysis of native gangliosides by MALDI-TOF. We identified the expression of GM1, GM2, GM3, GD2, GD3, GT2, and GT3 in SK-Mel28 (melanoma), LAN-1 (neuroblastoma), Hs 578T, SUM 159PT, MDA-MB-231, MCF-7 (BC), and BC cell lines over-expressing GD3 synthase. Among O-acetylated gangliosides, we characterized the expression of OAcGM1, OAcGD3, OAcGD2, OAcGT2, and OAcGT3. Furthermore, the experimental procedure allowed us to clearly identify the position of the sialic acid residue that carries the O-acetyl group on b- and c-series gangliosides by MS/MS fragmentation. These results show that ganglioside O-acetylation occurs on both inner and terminal sialic acid residue in a cell type-dependent manner, suggesting different O-acetylation pathways for gangliosides. They also highlight the limitation of immuno-detection for the complete identification of O-acetylated ganglioside profiles in cancer cells.

2019 ◽  
Vol 10 (1) ◽  
Author(s):  
Natalia Lemos Chaves ◽  
Danilo Aquino Amorim ◽  
Cláudio Afonso Pinho Lopes ◽  
Irina Estrela-Lopis ◽  
Julia Böttner ◽  
...  

Abstract Background Nanocarriers have the potential to improve the therapeutic index of currently available drugs by increasing drug efficacy, lowering drug toxicity and achieving steady-state therapeutic levels of drugs over an extended period. The association of maghemite nanoparticles (NPs) with rhodium citrate (forming the complex hereafter referred to as MRC) has the potential to increase the specificity of the cytotoxic action of the latter compound, since this nanocomposite can be guided or transported to a target by the use of an external magnetic field. However, the behavior of these nanoparticles for an extended time of exposure to breast cancer cells has not yet been explored, and nor has MRC cytotoxicity comparison in different cell lines been performed until now. In this work, the effects of MRC NPs on these cells were analyzed for up to 72 h of exposure, and we focused on comparing NPs’ therapeutic effectiveness in different cell lines to elect the most responsive model, while elucidating the underlying action mechanism. Results MRC complexes exhibited broad cytotoxicity on human tumor cells, mainly in the first 24 h. However, while MRC induced cytotoxicity in MDA-MB-231 in a time-dependent manner, progressively decreasing the required dose for significant reduction in cell viability at 48 and 72 h, MCF-7 appears to recover its viability after 48 h of exposure. The recovery of MCF-7 is possibly explained by a resistance mechanism mediated by PGP (P-glycoprotein) proteins, which increase in these cells after MRC treatment. Remaining viable tumor metastatic cells had the migration capacity reduced after treatment with MRC (24 h). Moreover, MRC treatment induced S phase arrest of the cell cycle. Conclusion MRC act at the nucleus, inhibiting DNA synthesis and proliferation and inducing cell death. These effects were verified in both tumor lines, but MDA-MB-231 cells seem to be more responsive to the effects of NPs. In addition, NPs may also disrupt the metastatic activity of remaining cells, by reducing their migratory capacity. Our results suggest that MRC nanoparticles are a promising nanomaterial that can provide a convenient route for tumor targeting and treatment, mainly in metastatic cells.


2020 ◽  
Vol 10 (3) ◽  
pp. 750 ◽  
Author(s):  
Megha Patel ◽  
Marek Feith ◽  
Birgit Janicke ◽  
Kersti Alm ◽  
Zahra El-Schich

Breast cancer is the second most common cancer type worldwide and breast cancer metastasis accounts for the majority of breast cancer-related deaths. Tumour cells produce increased levels of sialic acid (SA) that terminates the monosaccharide on glycan chains of the glycosylated proteins. SA can contribute to cellular recognition, cancer invasiveness and increase the metastatic potential of cancer cells. SA-templated molecularly imprinted polymers (MIPs) have been proposed as promising reporters for specific targeting of cancer cells when deployed in nanoparticle format. The sialic acid-molecularly imprinted polymers (SA-MIPs), which use SA for the generation of binding sites through which the nanoparticles can target and stain breast cancer cells, opens new strategies for efficient diagnostic tools. This study aims at monitoring the effects of SA-MIPs on morphology and motility of the epithelial type MCF-7 and the highly metastatic MDAMB231 breast cancer cell lines, using digital holographic cytometry (DHC). DHC is a label-free technique that is used in cell morphology studies of e.g., cell volume, area and thickness as well as in motility studies. Here, we show that MCF-7 cells move slower than MDAMB231 cells. We also show that SA-MIPs have an effect on cell morphology, motility and viability of both cell lines. In conclusion, by using DH microscopy, we could detect SA-MIPs impact on different breast cancer cells regarding morphology and motility.


2020 ◽  
Vol 2020 ◽  
pp. 1-14
Author(s):  
Stéphane Zingue ◽  
Abel Joël Gbaweng Yaya ◽  
Julia Cisilotto ◽  
Larissa Vanelle Kenmogne ◽  
Emmanuel Talla ◽  
...  

Abyssinone V-4′ methyl ether (AVME) isolated from Erythrina droogmansiana was recently reported to exhibit anti-mammary tumor effect in mice. The present work was therefore aimed at elucidating its cellular and molecular mechanisms. To achieve our goal, the cytotoxicity of AVME against tumoral and non-tumoral cell lines was evaluated by resazurin reduction test; flow cytometry allowed us to evaluate the cell cycle and mechanisms of cell death; the mitochondrial transmembrane potential, reactive oxygen species (ROS) levels, and caspase activities as well as apoptosis-regulatory proteins (Bcl-2 and Bcl-XL) were measured in MDA-MB-231 cells. Further, the antimetastatic potential of AVME was evaluated by invasion assay. AVME exhibited cytotoxic effects in all tested tumor cell lines and induced a significant increase in the percentage of MDA-MB-231 cells at G2/M and S phases of the cell cycle in a concentration-dependent manner. AVME also induced apoptosis in MDA-MB-231 cells, which was accompanied by the activation of caspase-3 and caspase-9 and downregulation of Bcl-2 and Bcl-XL proteins. Moreover, AVME suppressed cancer cell invasion by the inhibition of the metalloproteinase-9 activity. Findings from this study suggest that AVME has anti-breast cancer activities expressed through mitochondrial proapoptotic pathway including impairment of aggressive behaviors of breast cancer cells.


2020 ◽  
Vol 21 (21) ◽  
pp. 8282
Author(s):  
Elisabetta Mandarini ◽  
Eva Tollapi ◽  
Marta Zanchi ◽  
Lorenzo Depau ◽  
Alessandro Pini ◽  
...  

The process of heparan sulfate proteoglycan (HSPG) internalization has been described as following different pathways. The tumor-specific branched NT4 peptide has been demonstrated to bind HSPGs on the plasma membrane and to be internalized in tumor cell lines. The polycationic peptide has been also shown to impair migration of different cancer cell lines in 2D and 3D models. Our hypothesis was that HSPG endocytosis could affect two important phenomena of cancer development: cell migration and nourishment. Using NT4 as an experimental tool mimicking heparin-binding ligands, we studied endocytosis and trafficking of HSPGs in a triple-negative human breast cancer cell line, MDA-MB-231. The peptide entered cells employing caveolin- or clathrin-dependent endocytosis and macropinocytosis, in line with what is already known about HSPGs. NT4 then localized in early and late endosomes in a time-dependent manner. The peptide had a negative effect on CDC42-activation triggered by EGF. The effect can be explained if we consider NT4 a competitive inhibitor of EGF on HS that impairs the co-receptor activity of the proteoglycan, reducing EGFR activation. Reduction of the invasive migratory phenotype of MDA-MB-231 induced by NT4 can be ascribed to this effect. RhoA activation was damped by EGF in MDA-MB-231. Indeed, EGF reduced RhoA-GTP and NT4 did not interfere with this receptor-mediated signaling. On the other hand, the peptide alone determined a small but solid reduction in active RhoA in breast cancer cells. This result supports the observation of few other studies, showing direct activation of the GTPase through HSPG, not mediated by EGF/EGFR.


2012 ◽  
Vol 17 (7) ◽  
pp. 921-932 ◽  
Author(s):  
Neal Andruska ◽  
Chengjian Mao ◽  
Mathew Cherian ◽  
Chen Zhang ◽  
David J. Shapiro

Estrogens, acting through estrogen receptor α (ERα), stimulate breast cancer proliferation, making ERα an attractive drug target. Since 384-well format screens for inhibitors of proliferation can be challenging for some cells, inhibition of luciferase-based reporters is often used as a surrogate end point. To identify novel small-molecule inhibitors of 17β-estradiol (E2)–ERα-stimulated cell proliferation, we established a cell-based screen for inhibitors of E2-ERα induction of an estrogen response element (ERE)3–luciferase reporter. Seventy-five “hits” were evaluated in tiered follow-up assays to identify where hits failed to progress and evaluate their effectiveness as inhibitors of E2-ERα-induced proliferation of breast cancer cells. Only 8 of 75 hits from the luciferase screen inhibited estrogen-induced proliferation of ERα-positive MCF-7 and T47D cells but not control ERα-negative MDA-MB-231 cells. Although 12% of compounds inhibited E2-ERα-stimulated proliferation in only one of the ERα-positive cell lines, 40% of compounds were toxic and inhibited growth of all the cell lines, and ~37% exhibited little or no ability to inhibit E2-ERα-stimulated cell proliferation. Representative compounds were evaluated in more detail, and a lead ERα inhibitor was identified.


2007 ◽  
Vol 25 (18_suppl) ◽  
pp. 11105-11105
Author(s):  
A. Nakagawa ◽  
T. Sawada ◽  
T. Okada ◽  
T. Ohsawa ◽  
M. Adachi ◽  
...  

11105 Background: MK615 is an extract mixture from Japanese apricot, UME. In this study, the anti-neoplastic effects of MK615 against breast cancer cells were investigated. Methods: Two breast cancer cell lines, MDA-MB-468 (MDA) and MCF7, were cultured with (600, 300, 150 μg/ml) or without MK615. After 72 hours of incubation, growth inhibition was evaluated by MTT assay, and the mechanism of the anti-neoplastic effect of MK615 was evaluated by cell cycle- and apoptosis assay. Results: MK615 inhibited the growth of MDA and MCF7 in a dose-dependent manner. The percentage growth inhibition of MDA at dosages of 600, 300, and 150 μg/ml was 59.2%, 52.4%, and 23.3%, respectively, and that for MCF7 was 83.5%, 52.7%, and 16.6%, respectively. Cell cycle analysis showed that MK615 increased the proportion of cells in G2-M phase in both MDA (7.8% to 11.7%) and MCF7 (8.1% to 18.7%), and finally both cell lines became apoptotic. The proportion of apoptotic cells increased with incubation time. Conclusions: MK615 effectively inhibits the growth of breast cancer cells in vitro, possibly by cell cycle modification and apoptosis induction. No significant financial relationships to disclose.


2020 ◽  
Vol 16 (5) ◽  
pp. 726-733
Author(s):  
Guzide Satir Basaran ◽  
Hatice Bekci ◽  
Ayse Baldemir ◽  
Selen Ilgun ◽  
Ahmet Cumaoglu

Background and Objective: Herbal extracts and plant compounds are increasingly becoming of interest for their therapeutic potential in various cancer types. Momordica charantia is well known for its anti-diabetic, anti-inflammatory, and anti-cancer properties. Methods: In the present study, we investigated the antiproliferative and pro-apoptotic effects of Momordica charantia seed and aryl extracts on breast cancer cells and explored the underlying molecular mechanisms. Results: Our results showed that both extract significantly inhibited the growth of MCF-7 and MDA MB-231 cells in a concentration-dependent manner, and induced apoptosis by upregulation of caspase 9 and caspase 3 mRNA levels. In addition, in different incubation time, both extract evidently inhibited EGF and induced EGFR phosphorylation/activation in both cell lines. Moreover, Momordica charantia aryl and seed extracts inhibited phosphorylation/activation of PI3K/AKT and MAPK (ERK and P38) pathways in both cell lines. Conclusion: The current study clearly demonstrates that the Momordica charantia aryl and seed extracts have the potential to exert its cytotoxic effect on breast cancer cells by a mechanism involving inhibition of EGFR and EGRF related pathways with the induction of apoptosis. The overall finding demonstrates that this plant, especially seed extract, could be a potential source of new anticancer compounds for possible drug development against cancer.


2019 ◽  
Vol 20 (10) ◽  
pp. 804-814 ◽  
Author(s):  
Bing Wang ◽  
Yuzhu Zhang ◽  
Meina Ye ◽  
Jingjing Wu ◽  
Lina Ma ◽  
...  

Background: Chemoresistance blunts the therapeutic effect of cisplatin (DDP) on Triple-Negative Breast Cancer (TNBC). Researchers have not determined to date whether exosomes confer DDP resistance to other breast cancer cells or whether exosomal transfer of miRNAs derived from DDP-resistant TNBC cells confer DDP resistance. Objective: The aim of this study was to investigate the role of exosomes in chemoresistance in breast cancer. Methods: MDA-MB-231 cells resistant to DDP (231/DDP) were established. Exosomes were isolated from 231/DDP cells (DDP/EXO) and characterized by measuring the levels of protein markers, nanoparticle tracking analysis and transmission electron microscopy. MDA-MB-231, MCF-7 and SKBR-3 cell lines were treated with the isolated DDP/EXOs and cell proliferation and cytotoxicity to DDP were evaluated using MTT assays and apoptosis analyses. Western blotting was used to examine P-glycoprotein (P-gp) expression. Additionally, a microarray was used to analyse microRNA (miRNA) expression profiles in MDA-MB-231 and 231/DDP exosomes. The effects on miRNAs were determined using RT-PCR. Exosomal miR-423-5p was extracted, and differential expression was verified. The MTT cell viability assay, flow cytometry, and Transwell and immunofluorescence assays were performed to determine if differential expression of miR-423-5p sensitized cells to DDP in vitro. Results: Under a transmission electron microscope, the isolated exosomes exhibited a round or oval shape with a diameter ranging between 40 and 100 nm. DDP/EXOs labelled with PKH67 were taken up by MDA-MB-231 cells. After an incubation with DDP/EXOs, the cell lines exhibited a higher IC50 value for cisplatin, P-gp expression, migration and invasion capabilities and a lower apoptosis rate. Furthermore, 60 miRNAs from exosomes derived from 231/DDP cells were significantly up-regulated compared to exosomes from MDA-MB-231 cells. Notably, compared to the corresponding sensitive exosomes, miR-370-3p, miR-423-5p and miR-373 were the most differentially expressed miRNAs in DDP-resistant exosomes. We chose miR-423-5p, and up-regulation and down-regulation of exosomal miR-423-5p expression significantly affected DDP resistance. Conclusions: Exosomes from DDP-resistant TNBC cells (231/DDP) altered the sensitivity of other breast cancer cells to DDP in an exosomal miR-423-5p dependent manner. Our research helps to elucidate the mechanism of DDP resistance in breast cancer.


2021 ◽  
Author(s):  
Daniel Pereira Maurício de Barros ◽  
Ayla Nóbrega André ◽  
Basak Aru ◽  
Turkay Simsek ◽  
Gulderen Yanikkaya Demirel

Abstract With the emergence of Molecular Targeted Therapy, the interest in studying immunogenetic components that act in carcinogenesis has grown. The role of the estrogen receptor (ER) in initiation and progression of breast cancer is well documented and the estrogen treatment may affect expression of proteins described as tumor stem cell biomarkers in estrogen-sensitive breast cancer. The aim of this study is to analyze the expression of CD44 and CD326 on MCF-7 (ER+) and MDA-MB-231 (ER-) cell lines treated with 17β-estradiol for different periods. Our results indicate that 17β-estradiol can modulate CD44 and CD326 expression in breast cancer cells that have functional estrogen receptors in a time dependent manner. To our knowledge, this is the first study to investigate the influence of 17β-estradiol on CD44 and CD326 expression in MCF-7 and MDA-MB-231 cell lines. Further investigations with primary patient samples and their cultures will enhance our knowledge on the effect of hormones on breast cancer.


Sign in / Sign up

Export Citation Format

Share Document