scholarly journals Choriocarcinoma cell line Response to Dexamethasone

2010 ◽  
Vol 29 (2) ◽  
pp. 107-113
Author(s):  
Žanka Bojić-Trbojević ◽  
Nikola Kolundžić ◽  
Miloš Petronijević ◽  
Ljiljana Vićovac

Choriocarcinoma cell line Response to DexamethasoneChoriocarcinoma cell lines JAr and JEG-3 are model systems for the study of transformed trophoblast. Both cell lines were shown to produce galectin-1, expression of which was increased in choriocarcinoma when compared to the normal trophoblast of pregnancy. In this study the effects of synthetic glucocorticoid dexametha-sone were investigated in both JAr and JEG-3 cell lines by the MTT test, cell based ELISA, and the cell adhesion and migration tests. Viable cell number/cell proliferation of JAr cells was significantly increased after treatment with 0.1 and 1 nmol/L of dexamethasone, while proliferation of JEG-3 cells was significantly increased after treatment in the whole concentration range of dexamethasone (0.1-100 nmol/L). Galectin-1 in JAr cells was modulated by dexamethasone, which mildly, but significantly decreased production at low concentrations (0.1 and 1 nmol/L). In JEG-3 cells production of galectin-1 was significantly decreased only after treatment with 100 nmol/L of dexamethasone. Cell adhesion of JEG-3 was significantly increased in the presence of lactose, an inhibitory sugar for gal-1, while dexamethasone induced decrease of JEG-3 cell migration. These findings have shown that dexamet-hasone may affect proliferation, gal-1 production and cell migration, in a cell line specific manner. These data suggest that glucocorticoid treatmentin vivomight have the potential to affect cell functions in choriocarcinoma.

2005 ◽  
Vol 13 (1) ◽  
pp. 7-10 ◽  
Author(s):  
Zanka Bojic-Trbojevic ◽  
Miroslava Jankovic ◽  
Ljiljana Vicovac

BACKGROUND: JAr and Jeg-3 choriocarcinoma cell lines are model systems for the transformed trophoblast and allow studies of phenotype and regulatory factors for particular cell functions. Both cell lines express the receptor for insulin-like growth factor-I (IGF-I). Effects of IGF-I on adhesion, proliferation and galectin-1 production in JAr and Jeg-3 cells were studied. METHODS: The effects of IGF-I on proliferation and galectin-1 production were examined by thiazolyl blue assay and cell based solid phase assay using polyclonal anti-galectin-1 antibodies. The cell adhesion assay was performed on Matrigel coated wells. Galectin-1 production and localization was examined by immunocytochemistry. RESULTS: IGF-I decreased adhesion of JAr cells to 70% of the control value (p<0.05). Cell treatment with 10 ?g/L of IGF-I significantly increased viable cell number: by 13.5% in JAr and 6% in Jeg-3. Gal-1 was immunolocalized intracellularly and associated with the cell membrane in both cell lines. Production of galectin-1 was significantly increased after treatment with IGF-I compared to control: by 7% in JAr cells and by 16% in Jeg-3 cells (p<0.05). CONCLUSION: The data showed that IGF-I affected adhesion and proliferation of choriocarcinoma cells, depending on the cell line. Both choriocarcinoma cell lines studied here produced galectin-1. The amount of galectin-1 was moderately stimulated by IGF-I.


2021 ◽  
Author(s):  
Daniel Marinowic ◽  
Fabiana Spillari Viola ◽  
Fernanda Majolo ◽  
Gabriele Goulart Zanirati ◽  
Pamella Nunes Azevedo ◽  
...  

Abstract Glioblastoma (GBM) is one of the most common brain tumors in adults. Despite the presence of available treatments, it remains one of the most lethal and difficult tumors to treat such that most patients die within two years. Studies reported that infection with Zika virus (ZIKV) causes inhibition of cell proliferation as well as induction of apoptosis; moreover, these manifestations show a predilection for developing neuronal cells. In the present study, two GBM cell lines U-138 and U-251 were infected with ZIKV at multiplicities of infection (MOI) 0.1, 0,01 and 0.001 and tested for cell viability, cell migration, cell adhesion, induction of apoptosis, interleukin levels, and cell surface markers (CD14 and CD73). Our study demonstrated that the ZIKV infection promotes loss of cell viability and increased apoptosis potential. It was not evidenced changes in cell migration, however, the two glioblastoma cell lines displayed increased the cell adhesion behavior. There was small increase in the IL-4 level in the U-251 cell line after exposure to ZIKV, with no change in relation to INF-γ levels. Furthermore, we observed an increase in the percentage of cells expressing the CD14 surface marker in both cell lines and increased CD73 expression in the U-251 cell line. Our results suggest that ZIKV may be associated with decrease of cell viability and increased CD73 expression, enhanced adherence, as well as increased apoptosis rates. Further investigations are required to explore the potential use of ZIKV in the treatment of GBM.


1999 ◽  
Vol 112 (4) ◽  
pp. 579-587 ◽  
Author(s):  
D. Nath ◽  
P.M. Slocombe ◽  
P.E. Stephens ◽  
A. Warn ◽  
G.R. Hutchinson ◽  
...  

Metargidin (ADAM-15) is a type I transmembrane glycoprotein belonging to the ADAM (A Disintegrin and Metalloprotease Domain) family of proteins and is widely expressed in different tissues and cell types. Members of this family contain an amino-terminal metalloprotease domain followed by a disintegrin domain, a cysteine-rich region and a membrane proximal EGF-like domain. The disintegrin domain of metargidin contains an RGD tripeptide sequence, suggesting that it may potentially interact with the integrin family of proteins. Here we identify integrin ligands for metargidin on haemopoietic cells, by using a chimeric protein containing the extracellular domain of metargidin fused to the Fc portion of human IgG. Binding activity to a panel of human cell lines was analysed by solid-phase cell-adhesion assays. Metargidin bound to a monocytic cell line, U937, and a T cell line, MOLT-4, in a specific manner. Adhesion was divalent cation- and temperature- dependent and strongly enhanced by Mn2+, all features of integrin-mediated binding. Using a panel of anti-integrin antibodies we show that alphavbeta3 is a ligand for metargidin on U937 cells. In contrast, for MOLT-4 cells, the integrin alpha5beta1 contributes to cell binding. Adhesion was mediated by the disintegrin domain of metargidin as RGD-based peptides inhibited cell binding to both cell lines. The specificity of the interaction between both alphavbeta3 and alpha5beta1 and metargidin was further confirmed by solid-phase adhesion assays using purified recombinant integrins. These results together indicate that metargidin can function as a cell adhesion molecule via interactions with alphavbeta3 and alpha5beta1 integrins.


2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Rebecca Smith ◽  
Moqing Liu ◽  
Tiera Liby ◽  
Nora Bayani ◽  
Elmar Bucher ◽  
...  

AbstractRepresentative in vitro model systems that accurately model response to therapy and allow the identification of new targets are important for improving our treatment of prostate cancer. Here we describe molecular characterization and drug testing in a panel of 20 prostate cancer cell lines. The cell lines cluster into distinct subsets based on RNA expression, which is largely driven by functional Androgen Receptor (AR) expression. KLK3, the AR-responsive gene that encodes prostate specific antigen, shows the greatest variability in expression across the cell line panel. Other common prostate cancer associated genes such as TMPRSS2 and ERG show similar expression patterns. Copy number analysis demonstrates that many of the most commonly gained (including regions containing TERC and MYC) and lost regions (including regions containing TP53 and PTEN) that were identified in patient samples by the TCGA are mirrored in the prostate cancer cell lines. Assessment of response to the anti-androgen enzalutamide shows a distinct separation of responders and non-responders, predominantly related to status of wild-type AR. Surprisingly, several AR-null lines responded to enzalutamide. These AR-null, enzalutamide-responsive cells were characterized by high levels of expression of glucocorticoid receptor (GR) encoded by NR3C1. Treatment of these cells with the anti-GR agent mifepristone showed that they were more sensitive to this drug than enzalutamide, as were several of the enzalutamide non-responsive lines. This is consistent with several recent reports that suggest that GR expression is an alternative signaling mechanism that can bypass AR blockade. This study reinforces the utility of large cell line panels for the study of cancer and identifies several cell lines that represent ideal models to study AR-null cells that have upregulated GR to sustain growth.


2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Xuemei Li ◽  
Jike Hu ◽  
Baohong Gu ◽  
Maswikiti Ewetse Paul ◽  
Bofang Wang ◽  
...  

Abstract One of the most important and striking characteristics of hepatocellular carcinoma (HCC) with intrahepatic metastasis, is that it results in extremely poor prognosis. Animal models have become a fundamental and very useful in research for disease study. However, some limitation has arisen from these model systems. We have therefore established a model of HCC with intrahepatic metastasis and noticed some differential appearances in different HCC cell lines. Luciferase-transfected HCC cell lines MHCC97-H and PLC/PRF/5 were inoculated into SCID mice via spleen. Observation the intrahepatic metastasis by bioluminescence imaging in vivo and comparing of the differential formation of metastatic lesions between different HCC cell lines by incorporating physical anatomy was done. Animal models for HCC intrahepatic metastasis were well established. However, there were some clearly noticed differences between MHCC97-H and PLC/PRF/5 cell lines. The group of MHCC97-H cell line readily metastasis in the liver, whereas group PLC/PRF/5 cell line developed extensive intrahepatic metastasis and formed large tumor in situ in the spleen. MHCC97-H and PLC/PRF/5 cell lines can be used to successfully establish a model of HCC intrahepatic metastasis with distinctive characteristics, which provides an important direction for the study of the mechanism of HCC intrahepatic metastasis, and may hopefully provide a basis for clinical treatment.


2013 ◽  
Vol 94 (3) ◽  
pp. 497-506 ◽  
Author(s):  
Do Nyun Kim ◽  
Min Koo Seo ◽  
Hoyun Choi ◽  
Su Yeon Kim ◽  
Hee Jong Shin ◽  
...  

Epstein–Barr virus (EBV) is a herpesvirus associated with lymphomas and carcinomas. While EBV-associated epithelial cell lines are good model systems to investigate the role of EBV in carcinoma, only a few cell lines are available as they are hard to acquire. A greater variety of naturally EBV-infected cell lines which are derived from tumour patients are needed to represent various features of EBVaGC. We characterized cell line YCCEL1, established from a Korean EBVaGC patient, to ascertain whether it can be used to study the roles of EBV in EBVaGC. The expression of EBV genes and cell surface markers was examined by in situ hybridization, RT-PCR, Western blot analysis, immunofluorescence assay and Northern blot analysis. EBV episomal status was analysed by Southern blotting and real-time PCR. This cell line expressed EBV nuclear antigen 1 (EBNA1) and latent membrane protein 2A (LMP2A), but not EBNA2, LMP2B nor LMP1. The majority of the lytic proteins were not detected in YCCEL1 cells either before or after treatment with 12-O-tetradecanoylphorbol-13-acetate. YCCEL1 cells expressed BART microRNAs (miRNAs) at high level but did not express BHRF1 miRNAs. YCCEL1 cells expressed cytokeratin, but not CD21 and CD19, suggesting CD21-independent EBV infection. The latent EBV gene and EBV miRNA expression pattern of YCCEL1 cells closely resembled that of general EBVaGC cases. Our results support the value of YCCEL1 cells as a good model system to study the role of EBV in gastric carcinogenesis.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3305-3305 ◽  
Author(s):  
Matthew A. Kutny ◽  
Steven J. Collins ◽  
Keith Loeb ◽  
Roland B. Walter ◽  
Soheil Meshinchi

Abstract Abstract 3305 The differentiating agent ATRA has been used successfully in the treatment of acute promyelocytic leukemia (APL). By comparison, non-APL AML has not shown similar sensitivity to ATRA induced differentiation. Recent data has suggested that a subset of de novo AML patients with nucleophosmin (NPM1) mutations may benefit from addition of ATRA to conventional therapy. The NPM1 gene has several functions affecting cell cycle proliferation including regulation of ribosome biogenesis and centrosome duplication and it acts as a histone chaperone. Mutation of the NPM1 gene leads to differentiation arrest contributing to AML pathogenesis. We hypothesized that leukemia cells with NPM1 mutations could be induced to undergo differentiation. We tested this hypothesis with the NPM1 mutant AML cell line OCI-AML3 and compared the results to identical assays using the AML cell line HL-60 which has been previously well documented to differentiate in response to ATRA therapy. OCI-AML3 and HL-60 cell lines were treated for 5 days with control media and four ATRA doses including 0.2 μM, 1 μM, 5 μM, and 25 μM. Cell viability was assessed by flow cytometry. Compared to the control condition, OCI-AML3 cells treated with the lowest dose of ATRA (0.2 μM) had a live cell count 21.6% of the control. HL-60 cells treated at even the highest ATRA dose (25 uM) had a live cell count 79.3% of the control. Due to the sensitivity of OCI-AML3 cells to the toxic effects of ATRA, the experiment was repeated with lower doses of ATRA including 0.001 μM, 0.01 μM and 0.1 μM. At the lowest dose of ATRA (0.001 μM), OCI-AML3 cells demonstrated a cell viability of 49% with further decrease to 26% at 0.1 μM dose of ATRA. At similar ATRA doses, cell viability for HL-60 cells was 91% and 85%, respectively (see table 1). Table 1: Cell viability as a percent of control cells after 5 days of treatment at three different doses of ATRA in OCI-AML3 and HL-60 cell lines. Cell Line: ATRA 0.001 μM ATRA 0.01 μM ATRA 0.1 μM OCI-AML3 49% 33% 26% HL-60 91% 91% 85% We subsequently determined the time course of changes in cell growth and the extent of differentiation at each point was determined by morphologic assessment. Both cell lines were treated with ATRA at doses of 0.001 μM, 0.01 μM, 0.1 μM, and 1 μM for a total of 4 days. Each day viable cell number was determined. In contrast to the HL-60 cells which had continued growth in lower ATRA doses, OCI-AML3 cells demonstrated exquisite sensitivity to growth arrest at the lowest doses of ATRA. Cell morphology was assessed daily with modified Wright-Giemsa staining of cells. Cells were examined for signs of myeloid differentiation including decrease in nuclear to cytoplasmic (N/C) ratio, nuclear segmentation, and cytoplasmic granules and vacuoles. At the lowest dose of ATRA (0.001 μM), after 4 days of exposure, significant number of OCI-AML3 cells demonstrated morphologic evidence of differentiation. At this ATRA dose and exposure interval, HL-60 cells showed no evidence of differentiation. At an ATRA dose of 1 μM (considered a standard dose used for differentiation of HL-60 cells), the OCI-AML3 cells showed differentiation changes as early as day 2 with nuclear segmentation and decreased N/C ratio while HL-60 cells did not show any change at this time point. After 4 days of ATRA exposure, most OCI-AML3 cells showed segmented nuclei and vacuolated cytoplasm, whereas HL-60 cells showed less distinct signs of differentiation with some cytoplasm granules and cup shaped nuclei. This data suggests that leukemic cells with NPM mutations may be susceptible to the pro-differentiating properties of ATRA. Further substantiation of this data with primary human specimens may ultimately provide the rationale for a novel therapeutic option using ATRA-based differentiation therapy for subsets of non-APL AML. Disclosures: No relevant conflicts of interest to declare.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. e13563-e13563
Author(s):  
Andre P. Fay

e13563 Background: Glioblastoma (GBM) is the most common central nervous system (CNS) tumor. Despite available therapies survival remains poor, and new treatment strategies are needed. Oncolytic viral therapy is under investigation in GBM. Zika virus (ZV) infection has demonstrated inhibition of neuronal precursor cells proliferation in preclinical studies. This study aims to evaluate the effects of ZV infection on human glioblastoma cell lines survival. Methods: Two GBM cell lines (U138 and U 251) were infected by 2 hours with ZV using the titer of PFU/ml diluted in 1:100, 1:1000, 1: 10000. Cell. We performed tests to evaluate cell viability by MTT and protease active assay, cell migration trough a wound healing assay and cell infiltration using insert culture method. Also the cell lines were tested by cell adhesion capacity, apoptosis (Caspase 3/7), interleukins levels and cell surface markers for CD 14 and CD73.These results were compared to controls. Results: Our study has demonstrated a reduction in cell viability in U138 lineage trough MTT assay. In the U251 lineage and in the others tests for cytotoxicity/viability, ZK did not altered cell viability neither cell migration compared to controls. We showed that ZV caused reduction in cell invasion and resulted in increased rates of apoptosis in both cell lines. We observed that infection of ZV caused increased rates of cell adhesion and CD73 marker. Conclusions: These findings suggest that ZV infection may be associated with increased CD 73 Expression thus increasing cell adhesion and cell infiltration. The ZV may be cause an increase rates of apoptosis and influencing cell cytotoxicity and viability. Further investigations to explore the role of ZV in GBM treatment are warranted.


2017 ◽  
Vol 37 (2) ◽  
Author(s):  
Run-Tian Liu ◽  
Jing-Lin Cao ◽  
Chang-Qing Yan ◽  
Yang Wang ◽  
Cong-Jing An ◽  
...  

The present study explored the effect of long non-coding RNA-human ovarian cancer-specific transcript 2 (LncRNA-HOST2) on cell proliferation, migration, invasion and apoptosis of human hepatocellular carcinoma (HCC) cell line SMMC-7721. HCC tissues and adjacent normal tissues from 162 HCC patients were collected. The HCC cell lines were assigned into the control group (regular culture), negative control (NC) group (transfected with siRNA) and experimental group (transfected with Lnc-HOST2 siRNA). Quantitative real-time PCR (qRT-PCR) was used to detect the expression of LncRNA-HOST2. Cell proliferation was detected by CCK-8 and colony-forming assays, cell apoptosis by flow cytometry and cell migration by Scratch test. Transwell assay was used to evaluate cell migration and invasion abilities. LncRNA-HOST2 expression in the HCC tissues increased 2–10 times than that in the adjacent normal tissues. Compared with the HL-7702 cell line, LncRNA-HOST2 expression in HepG2, SMMC-7721 and Huh7 cell lines was all up-regulated, but the SMMC-7721 cell had the highest Lnc-HOST2 expression. The LncRNA-HOST2 expression in the experimental group was down-regulated as compared with the control and NC groups. In comparison with the control and NC groups, cloned cells reduced, cell apoptosis increased, clone-forming ability weakened and inhibitory rate of colony formation increased in the experimental group. The cells migrating and penetrating into the transwell chamber were fewer in the experimental group than those in the control and NC groups. The experimental group exhibited slow wound healing and decreased cell migration area after 48 h. These findings indicate that LncRNA-HOST2 can promote cell proliferation, migration and invasion and inhibit cell apoptosis in human HCC cell line SMMC-7721.


2019 ◽  
Author(s):  
Rupam Bhattacharyya ◽  
Min Jin Ha ◽  
Qingzhi Liu ◽  
Rehan Akbani ◽  
Han Liang ◽  
...  

ABSTRACTPurposePersonalized network inference on diverse clinical and in vitro model systems across cancer types can be used to delineate specific regulatory mechanisms, uncover drug targets and pathways, and develop individualized predictive models in cancer.Datasets and methodsWe developed TransPRECISE, a multi-scale Bayesian network modeling framework, to analyze the pan-cancer patient and cell line interactome to identify differential and conserved intra-pathway activities, globally assess cell lines as representative models for patients and develop drug sensitivity prediction models. We assessed pan-cancer pathway activities for a large cohort of patient samples (>7700) from The Cancer Proteome Atlas across ≥30 tumor types and a set of 640 cancer cell lines from the M.D. Anderson Cell Lines Project spanning16 lineages, and ≥250 cell lines’ response to >400 drugs.ResultsTransPRECISE captured differential and conserved proteomic network topologies and pathway circuitry between multiple patient and cell line lineages: ovarian and kidney cancers shared high levels of connectivity in the hormone receptor and receptor tyrosine kinase pathways, respectively, between the two model systems. Our tumor stratification approach found distinct clinical subtypes of the patients represented by different sets of cell lines: head and neck patient tumors were classified into two different subtypes that are represented by head and neck and esophagus cell lines, and had different prognostic patterns (456 vs. 654 days of median overall survival; P=0.02). The TransPRECISE-based sample-specific pathway scores achieved high predictive accuracy for drug sensitivities in cell lines across multiple drugs (median AUC >0.8).ConclusionOur study provides a generalizable analytical framework to assess the translational potential of preclinical model systems and guide pathway-based personalized medical decision-making, integrating genomic and molecular data across model systems.


Sign in / Sign up

Export Citation Format

Share Document