scholarly journals Combinatorial Analysis of AT-Rich Interaction Domain 1A and CD47 in Gastric Cancer Patients Reveals Markers of Prognosis

Author(s):  
Qianfu Zhao ◽  
Qu Cai ◽  
Shanhe Yu ◽  
Jun Ji ◽  
Zhenggang Zhu ◽  
...  

Background: The AT-rich interaction domain 1A (ARID1A) is thought to be a tumor suppressive gene, and most of its mutations result in loss of expression of ARID1A protein. Combined with SIRPα on the surface of macrophages, CD47 on the surface of cancer cells can send an antiphagocytic “Don’t eat me” signal to the immune system that helps to avoid immune surveillance. However, the relationship between ARID1A and CD47 expression and their prognostic value in gastric cancer (GC) are still unknown.Methods: In this study, we evaluated ARID1A and CD47 expression in 154 GC patients’ tissues using tissue microarray. Expressions of ARID1A and CD47 in GC cell lines were determined by western blot and quantitative reverse transcriptase–polymerase chain reaction (qRT-PCR) techniques, and cell membranous CD47 expression was quantified by flow cytometry. In addition, chromatin immunoprecipitation (ChIP)–qPCR was used to determine the aspects of regulation of CD47 by ARID1A. The proportions of tumor-infiltrating immune cells were estimated on The Cancer Genome Atlas (TCGA) data set by using quanTIseq and EPIC algorithms. The infiltration of M1-polarized macrophages, M2-polarized macrophages, and regulatory T cells (Tregs) in GC tissues was determined by multispectral immunofluorescence.Results: A significant correlation was found between loss of ARID1A and high expression of CD47 at protein level in GC. By integrating 375 bulk RNA sequencing samples from TCGA data set, we found that mutated ARID1A correlated with high CD47 expression. In GC cell lines, knockdown of ARID1A significantly increased CD47 expression both at protein and mRNA levels as measured by western blot, qRT-PCR, and flow cytometry. Moreover, ChIP-qPCR revealed that CD47 was a direct downstream target gene of ARID1A in GC. Utilizing univariate and multivariate survival analyses, we found that patients with ARID1AlossCD47high expression had a worse prognosis. Estimation of infiltrating immune cells on TCGA data set showed that a higher infiltration proportion of M2 macrophages and Tregs was found in ARID1AmutatedCD47high expression subgroup. Furthermore, application of multispectral immunofluorescence revealed a higher infiltration proportion of M2 macrophages and Tregs in ARID1AlossCD47high GC tissues.Conclusion: Loss of ARID1A is strongly correlated with high CD47 expression in GC, and combination of ARID1A and CD47 is a promising prognosis factor in GC.

2020 ◽  
Author(s):  
LU GE ◽  
Chang-long Hu ◽  
Zheng-hui Ge ◽  
Chun-rong Wang ◽  
Li Qian ◽  
...  

Abstract Purpose Capicua homolog protein (CIC) played a broad role in the development of cancer in humans, however, its role in the progression of gastric cancer (GC) specifically has been unclear. This study aimed to explore the expression of CIC and its potential clinical value in patients with GC. Methods The CIC levels in GC tissues and cell lines were examined by quantitative real-time polymerase chain reaction (qRT-PCR). And the in-vitro effects of CIC expression in MGC-803 cells on their proliferation, invasion, and the progression of epithelial-mesenchymal transition were assessed by CCK-8 assays, Matrigel-invasion analysis, qRT-PCR and Western blot assays, separately. In addition, the effects of downregulation of CIC on the activation of PI3K/AKT signaling pathway were measured using Western-blot analysis. Results The results showed CIC levels were lower in GC tissues and GC cell lines, and these lower CIC levels were correlated with tumor differentiation, Helicobacter pylori infection, TNM stage, and patient survival. In addition, CIC overexpression could promote cell proliferation, invasion, and progression of epithelial-mesenchymal transition in MGC-803 cells. Notably, exotic expression of CIC inactivated the phosphoinositide 3-kinase/protein kinase B signaling pathway. Conclusions In conclusion, our finding suggested CIC could serve as a potential diagnostic and prognostic biomarker and a probable therapy target for GC.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2086-2086
Author(s):  
Xing Guo ◽  
Donghua He ◽  
Jing Chen ◽  
Xuanru Lin ◽  
Qingxiao Chen ◽  
...  

Abstract Background: Multiple myeloma (MM) is the second mostly diagnosed disease among hematological malignancies after lymphoma. With the novel agents, the survival of MM patients has been improved significantly but still remains incurable because of drug resistance. Studies have found that high-mobility group box 1 protein (HMGB1) was involved in inflammation, angiogenesis, and cancer invasion progression, metastasis, and drug resistance. Our research was aimed at exploring the role of HMGB1 in MM cell proliferation and drug resistance. Methods: First, semi-quantitative real time-polymerase chain (qRT-PCR) and western blot was used to determine the levels of HMGB1 mRNA and protein expression in MM cell lines (RPMI8226, CAG, and MM.1S) and primary MM samples. Second, MM cells were transfected with HMGB1-knockdown lentivirus and the Cell Counting Kit 8 (CCK8) assay was used to determine the proliferation of MM cells with or without chemotherapeutic drugs dexamethasone (Dex) and doxorubicin (ADM). Then cell apoptosis was detected by flow cytometry. Third, Affymetrix HTA 2.0 Array was used to compare changes in gene expression levels between HMGB1-knockdown cells and the control cells and qRT-PCR was used to verify the array results. Last, Western bolt was performed to analyze changes in signaling pathways after HMGB1 knockdown. Results: MM cell lines and primary MM samples expressed high levels of HMGB1 mRNA and protein. Although there was no difference in MM cell proliferation between HMGB1-knockdown group and the control group (P>0.05), HMGB1-knockdown significantly enhanced inhibitory effect of chemotherapy with Dex and ADM in comparison with the wildtype HMGB1 control (P<0.05). Flow cytometry results showed that apoptosis of MM cells induced by Dex and ADM was increased when HMGB1 expression was suppressed (P<0.05). Furthermore, gene array analysis on RPMI8226 and CAG cell lines showed that anti-apoptotic genes (bcl-2, HIAP1) and MM survival related genes (DEPTOR, CXCL12) were decreased and pro-apoptotic genes (TNFRSF1B, TRAIL, CXCL10) were increased in knockdown cells compared to the controls. In addition, expression levels of genes that play important roles in signaling pathways such as mTOR, NF-κB, PI3K-AKt, and p38-MAPK were also significantly changed. The gene expression microarray results were verified by qRT-PCR and Western blot demonstrated that phosphorylation of p70S6K (substrate of mTORC1 complex) and AKT-ser473 (substrate of mTORC2 complex) were both elevated in HMGB1-knockdown MM cells compared to that in the control cells. Conclusions: Our research showed that downregulation of HMGB1 increased sensitivity of MM cells to Dex and ADM through increasing apoptosis and regulating mTOR, NF-κB, PI3K-AKt, and p38-MAPK pathways. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Author(s):  
Jinshan Liu ◽  
Ximing Huang ◽  
Liran Wu ◽  
Zhiqiang Zhao ◽  
Kun Fan

Abstract Objective: MiR-498 has emerged as a potential molecular target for several cancer. In this study, we aimed to investigate the important function and mechanisms of miR-498 in gastric cancer.Methods: To detect the important roles of miR-498 in gastric cancer, we first measured its expression by RT-qPCR in gastric cancer cell lines. The impact of the miR-498 on gastric cancer cell proliferation were detected by CCK-8 and colony formation assays. The effect of the miR-498 on the cell apoptosis and cell cycle were detected by flow cytometry. We also used the scratch and transwell chamber assays to measure the cell migration and invasion. The expression levels of related proteins were assessed by western blot. The bioinformatics analysis was used to explore the target gene of miR-498. RT-qPCR and western blot assays were used to detect the expression levels of FOXK1 in response to miR-498 overexpression. In order to prove the role of FOXK1 in mediating the effect of miR-498 on the gastric cancer, CCK-8, colony formation, transwell chamber and flow cytometry assays were used for the further investigations.Results: The expression level of miR-498 is downregulated in gastric cancer cell lines. Overexpression of miR-498 inhibited proliferation and migration/invasion, while promoted the apoptosis of gastric cancer cells. Bioinformatics analysis indicated that miR-498 targeted on FOXK1 to inhibit the gastric cancer in vitro.Conclusion: MiR-498/FOXK1 axis may be a potential therapeutic target for the treatment of gastric cancers.


2020 ◽  
Vol 79 (Suppl 1) ◽  
pp. 573.1-573
Author(s):  
L. Ehlers ◽  
A. Kuppe ◽  
A. Damerau ◽  
M. Kirchner ◽  
C. Strehl ◽  
...  

Background:Adenosine and its nucleotides represent crucial immunomodulators in the extracellular environment. ATP and ADP are released from stressed cells in states of inflammation, whereas adenosine serves as a key anti-inflammatory mediator1. The ectonucleotidases CD39 and CD73 are responsible for the sequential catabolism of ATP to adenosine via AMP, thereby promoting an anti-inflammatory milieu induced by the “adenosine halo”. Great importance has been attributed to these enzymes in the pathogenesis of autoimmune diseases such as rheumatoid arthritis (RA) and as targets in cancer therapy2 3. AMPD2 mediates AMP deamination to IMP, thus constituting an ambiguous mediator both enhancing the degradation of inflammatory ATP and reducing the formation of protective adenosine. Here, we postulate that this pathway is also present on the cell surface of immune cells and modified under inflammatory conditions.Objectives:Therefore, we analysed surface AMPD2 expression and its modulation on distinct cell lines and primary immune cells.Figure 1.Surface AMPD2 as a novel regulator of the extracellular ATP-adenosine balance.Methods:Firstly, AMPD2 surface expression was verified by immunoprecipitation from membrane fractions isolated from cell lines (HEK293 and HMEC1) and CD14+ monocytes analysed by western blot and mass spectrometry. In addition, surface biotinylation of the aforementioned cells was performed. Also, AMPD2 surface expression was evaluated by flow cytometry, analysing both cell lines (HEK293, HMEC1, THP1, and Jurkat) and primary human immune cells from healthy donors and patients with RA.Secondly, co-expression of surface AMPD2, CD39 and CD73 on PBMCs was analysed by flow cytometry directly after isolation as well as after a 24h culture period. Moreover, surface expression was assessed after immunostimulation and Golgi transport inhibition.Results:AMPD2 surface expression was confirmed by western blot and mass spectrometry of (i) precipitated AMPD2 from membrane fractions and (ii) biotinylated surface molecules in HEK293 and HMEC1 as well as CD14+ monocytes. Surface expression was reduced after AMPD2 knockdown in HEK293. Flow cytometric analysis further verified AMPD2 surface expression and revealed a significant decrease after Golgi transport inhibition (p<0.01). TLR stimulation strongly enhanced the surface expression of AMPD2 and CD39 on monocytes (p<0.05), whereas dexamethasone at high therapeutic doses inversely affected AMPD2 surface expression on lymphocytes and monocytes (p<0.01). Analysis of AMPD2 surface expression on PBMCs from RA patients revealed higher expression levels compared to sex- and age-matched healthy controls (p<0.05).Conclusion:We demonstrate AMPD2 surface expression on immune cells for the first time. Hence, we reveal a novel regulator of the extracellular ATP-adenosine balance that is differentially expressed in RA patients compared to healthy controls. The extracellular conversion of AMP into IMP may constitute a shunt-like mechanism adding to the CD39-CD73 system controlling immunomodulation.References:[1]Regateiro FS, Cobbold SP, Waldmann H. CD73 and adenosine generation in the creation of regulatory microenvironments.Clinical and experimental immunology2013;171(1):1-7. doi: 10.1111/j.1365-2249.2012.04623.x[2]Morandi F, Horenstein AL, Rizzo R, et al. The Role of Extracellular Adenosine Generation in the Development of Autoimmune Diseases.Mediators of inflammation2018;2018:7019398. doi: 10.1155/2018/7019398[3]Allard B, Longhi MS, Robson SC, et al. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets.Immunol Rev2017;276(1):121-44. doi: 10.1111/imr.12528Acknowledgments:This project is funded by an unrestricted grant by Horizon Pharma plc.Disclosure of Interests:Lisa Ehlers: None declared, Aditi Kuppe: None declared, Alexandra Damerau: None declared, Marieluise Kirchner: None declared, Cindy Strehl: None declared, Frank Buttgereit Grant/research support from: Amgen, BMS, Celgene, Generic Assays, GSK, Hexal, Horizon, Lilly, medac, Mundipharma, Novartis, Pfizer, Roche, and Sanofi., Timo Gaber: None declared


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Linwen Zhu ◽  
Zhe Li ◽  
Xiuchong Yu ◽  
Yao Ruan ◽  
Yijing Shen ◽  
...  

Abstract Background Recently, tRNA-derived fragments (tRFs) have been shown to serve important biological functions. However, the role of tRFs in gastric cancer has not been fully elucidated. This study aimed to identify the tumor suppressor role of tRF-5026a (tRF-18-79MP9P04) in gastric cancer. Methods Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was first used to detect tRF-5026a expression levels in gastric cancer tissues and patient plasma. Next, the relationship between tRF-5026a levels and clinicopathological features in gastric cancer patients was assessed. Cell lines with varying tRF-5026a levels were assessed by measuring tRF-5026a using qRT-PCR. After transfecting cell lines with a tRF-5026a mimic or inhibitor, cell proliferation, colony formation, migration, apoptosis, and cell cycle were evaluated. The expression levels of related proteins in the PTEN/PI3K/AKT pathway were also analyzed by Western blotting. Finally, the effect of tRF-5026a on tumor growth was tested using subcutaneous tumor models in nude mice. Results tRF-5026a was downregulated in gastric cancer patient tissues and plasma samples. tRF-5026a levels were closely related to tumor size, had a certain diagnostic value, and could be used to predict overall survival. tRF-5026a was also downregulated in gastric cancer cell lines. tRF-5026a inhibited the proliferation, migration, and cell cycle progression of gastric cancer cells by regulating the PTEN/PI3K/AKT signaling pathway. Animal experiments showed that upregulation of tRF-5026a effectively inhibited tumor growth. Conclusions tRF-5026a (tRF-18-79MP9P04) is a promising biomarker for gastric cancer diagnostics and has tumor suppressor effects mediated through the PTEN/PI3K/AKT signaling pathway.


PLoS ONE ◽  
2021 ◽  
Vol 16 (1) ◽  
pp. e0246197
Author(s):  
Jorge Marquez ◽  
Jianping Dong ◽  
Chun Dong ◽  
Changsheng Tian ◽  
Ginette Serrero

Antibody-drug conjugates (ADC) are effective antibody-based therapeutics for hematopoietic and lymphoid tumors. However, there is need to identify new targets for ADCs, particularly for solid tumors and cancers with unmet needs. From a hybridoma library developed against cancer cells, we selected the mouse monoclonal antibody 33B7, which was able to bind to, and internalize, cancer cell lines. This antibody was used for identification of the target by immunoprecipitation and mass spectrometric analysis, followed by target validation. After target validation, 33B7 binding and target positivity were tested by flow cytometry and western blot analysis in several cancer cell lines. The ability of 33B7 conjugated to saporin to inhibit in vitro proliferation of PTFRN positive cell lines was investigated, as well as the 33B7 ADC in vivo effect on tumor growth in athymic mice. All flow cytometry and in vitro internalization assays were analyzed for statistical significance using a Welsh’s T-test. Animal studies were analyzed using Two-Way Analysis of Variance (ANOVA) utilizing post-hoc Bonferroni analysis, and/or Mixed Effects analysis. The 33B7 cell surface target was identified as Prostaglandin F2 Receptor Negative Regulator (PTGFRN), a transmembrane protein in the Tetraspanin family. This target was confirmed by showing that PTGFRN-expressing cells bound and internalized 33B7, compared to PTGFRN negative cells. Cells able to bind 33B7 were PTGFRN-positive by Western blot analysis. In vitro treatment PTGFRN-positive cancer cell lines with the 33B7-saporin ADC inhibited their proliferation in a dose-dependent fashion. 33B7 conjugated to saporin was also able to block tumor growth in vivo in mouse xenografts when compared to a control ADC. These findings show that screening antibody libraries for internalizing antibodies in cancer cell lines is a good approach to identify new cancer targets for ADC development. These results suggest PTGFRN is a possible therapeutic target via antibody-based approach for certain cancers.


2018 ◽  
Vol 132 (9) ◽  
pp. 1003-1019 ◽  
Author(s):  
Zihao Chen ◽  
Hongping Ju ◽  
Shan Yu ◽  
Ting Zhao ◽  
Xiaojie Jing ◽  
...  

Gastric cancer (GC) is one of the major global health problems, especially in Asia. Nowadays, long non-coding RNA (lncRNA) has gained significant attention in the current research climate such as carcinogenesis. This research desires to explore the mechanism of Prader–Willi region non-protein coding RNA 1 (PWRN1) on regulating GC process. Differentially expressed lncRNAs in GC tissues were screened out through microarray analysis. The RNA and protein expression level were detected by quantitative real-time PCR (qRT-PCR) and Western blot. Cell proliferation, apoptosis rate, metastasis abilities were respectively determined by cell counting kit 8 (CCK8), flow cytometry, wound healing, and transwell assay. The luciferase reporter system was used to verify the targetting relationships between PWRN1, miR-425-5p, and phosphatase and tensin homolog (PTEN). RNA-binding protein immunoprecipitation (RIP) assay was performed to prove whether PWRN1 acted as a competitive endogenous RNA (ceRNA) of miR-425-5p. Tumor xenograft model and immunohistochemistry (IHC) were developed to study the influence of PWRN1 on tumor growth in vivo. Microarray analysis determined that PWRN1 was differently expressed between GC tissues and adjacent tissues. qRT-PCR revealed PWRN1 low expression in GC tissues and cells. Up-regulated PWRN1 could reduce proliferation and metastasis and increase apoptosis in GC cells, while miR-425-5p had reverse effects. The RIP assay indicated that PWRN1 may target an oncogene, miR-425-5p. The tumor xenograft assay found that up-regulated PWRN1 suppressed the tumor growth. The bioinformatics analysis, luciferase assay, and Western blot indicated that PWRN1 affected PTEN/Akt/MDM2/p53 axis via suppressing miR-425-5p. Our findings suggested that PWRN1 functioned as a ceRNA targetting miR-425-5p and suppressed GC development via p53 signaling pathway.


2018 ◽  
Vol 49 (6) ◽  
pp. 2151-2162 ◽  
Author(s):  
Bo Lian ◽  
Dongxiang Yang ◽  
Yanlong Liu ◽  
Gang Shi ◽  
Jibin Li ◽  
...  

Background/Aims: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an ideal anti-tumor drug because it exhibits selective cytotoxicity against cancer cells. However, certain cancer cells are resistant to TRAIL, and the potential mechanisms are still unclear. The aim of this study was to reduce the resistance of colorectal cancer (CRC) cells to TRAIL. Methods: Quantitative real-time PCR analysis was performed to detect the expression of microRNA-128 (miR-128) in tissues from patients with CRC and CRC cell lines. MTT assays were used to evaluate the effect of miR-128 on TRAIL-induced cytotoxicity against CRC cell lines. The distribution of death receptor 5 (DR5) and the production of reactive oxygen species (ROS) were detected by flow cytometry analysis. Western blot, flow cytometry, and luciferase reporter assays were performed to evaluate the potential mechanism and pathway of miR-128-promoted apoptosis in TRAIL-treated CRC cells. Results: MiR-128 expression was downregulated in tumor tissues from patients with CRC as well as in CRC cell lines in vitro. The enforced expression of miR-128 sensitized CRC cells to TRAIL-induced cytotoxicity by inducing apoptosis. Mechanistically, bioinformatics, western blot analysis, and luciferase reporter assays showed that miR-128 directly targeted sirtuin 1 (SIRT1) in CRC cells. miR-128 overexpression suppressed SIRT1 expression, which promoted the production of ROS in TRAIL-treated CRC cells. This increase of ROS subsequently induced DR5 expression, and thus increased TRAIL-induced apoptosis in CRC cells. Conclusion: The combination of miR-128 with TRAIL may represent a novel approach for the treatment of CRC.


2020 ◽  
Vol 2020 ◽  
pp. 1-10
Author(s):  
Gang Wang ◽  
Dan Sun ◽  
Wenhui Li ◽  
Yan Xin

The AT-hook transcription factor, AKNA, is a nuclear protein that affects a few physiological and pathological processes including cancer. Here, we investigated the role of AKNA in gastric cancer (GC). By using quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot assays, AKNA was found deregulated in both GC cell lines and 32 paired GC tissues. Subsequently, Kaplan-Meier analysis and clinicopathological analysis were conducted using both 32 GC cases’ data above and RNA-Seq data of AKNA in 354 GC patients and the corresponding clinical-pathological data obtained from The Cancer Genome Atlas (TCGA), and AKNA expression was found closely related to location, metastasis, and TNM staging of GC. Then, the potential molecular mechanisms of AKNA in GC were explored by gene set enrichment analysis (GSEA), qRT-PCR, and Western blot assays. AKNA was found to be a hub gene related to homotypic cell to cell adhesion, regulation of cell to cell adhesion, leukocyte cell to cell adhesion, and regulation of T cell proliferation in GC. GO analysis revealed that AKNA involved in the regulation of epithelial-mesenchymal transition (EMT)-related pathways including chemokine signaling pathway, cytokine to cytokine receptor interaction, cell adhesion molecules, and jak-stat signaling pathway in GC. To explore the regulation of AKNA expression, Targetscan and TargetMiner were used to predict the possible miRNA which targeted AKNA and found the expression of AKNA was negatively correlated to miR-762 which could be sponged by circTRNC18. In conclusion, AKNA could function as a tumor suppressor by modulating EMT-related pathways in GC. The expression of AKNA might be regulated by circTRNC18/miR-762 axis. AKNA could serve as a potential biomarker and an effective target for GC diagnosis and therapy.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2904-2904
Author(s):  
Robyn M. Dennis ◽  
Brandon Ballard ◽  
David John Tweardy ◽  
Karen Rabin

Abstract Abstract 2904 Survival has improved dramatically in acute lymphoblastic leukemia (ALL), but further gains are unlikely using conventional chemotherapy alone. Several recently discovered, novel cytogenetic lesions with adverse prognostic impact, JAK2 activating mutations and CRLF2 rearrangements, occur in up to 15% of adult and pediatric ALL. These lesions are associated with activation of Jak2 and Stat5, and hold promise as targets for novel therapies affecting these signaling pathways. We performed in vitro testing of a novel small molecule Stat inhibitor, C188-9, in B-lineage ALL cell lines and patient samples with and without JAK2/CRLF2 alterations. C188-9 treatment for one hour at 10 μM inhibited Stat3 and Stat5 phosphorylation in ALL cell lines with JAK2 and CRLF2 alterations, but not in cell lines with wild-type JAK2 and CRLF2, as measured by phospho-flow cytometry (Fig. 1A). Only the cell lines with JAK2 and CRLF2 alterations demonstrated basal Stat5 phosphorylation on Western blot analysis, and this was inhibited by C188-9 treatment (Fig. 1B). C188-9 demonstrated cytotoxicity in ALL cell lines regardless of JAK2/CRLF2 status, with IC50s in the low micromolar concentration range (Fig. 1C). While C188-9 is undergoing investigation currently as a potent inhibitor of Stat3 in acute myeloid leukemia (AML), it also merits further investigation as an agent with Stat5 inhibitory activity and cytotoxicity in ALL. Figure 1. Effects of C188-9 in ALL cell lines. A. Stat3 and Stat5 phosphorylation were determined by flow cytometry in the ALL cell lines MHH-CALL-4 (JAK2/CRLF2 mutated) and Reh (JAK2/CRLF2 wild-type). In each condition, cells were incubated in serum-free media for one hour, followed by incubation with C188-9 or vehicle for one hour, stimulation with vehicle or pervanadate 125 mM for 15 minutes, fixation, permeabilization, phospho-antibody staining for phospho-Stat3 and phospho-Stat5, and flow cytometric analysis. B. Western blot for phospho-Stat5 in K562 cell line (positive control); MHHCALL-4 treated for one hour with C188-9 at 0, 5, or 10 uM; and RS4;11 (JAK2/CRLF2 wild-type ALL cell line). C. IC50 determination by ATP assay for C188-9 in the ALL cell lines MHH-CALL-4 and RS4;11. Each experiment was performed in triplicate. Figure 1. Effects of C188-9 in ALL cell lines. A. Stat3 and Stat5 phosphorylation were determined by flow cytometry in the ALL cell lines MHH-CALL-4 (JAK2/CRLF2 mutated) and Reh (JAK2/CRLF2 wild-type). In each condition, cells were incubated in serum-free media for one hour, followed by incubation with C188-9 or vehicle for one hour, stimulation with vehicle or pervanadate 125 mM for 15 minutes, fixation, permeabilization, phospho-antibody staining for phospho-Stat3 and phospho-Stat5, and flow cytometric analysis. B. Western blot for phospho-Stat5 in K562 cell line (positive control); MHHCALL-4 treated for one hour with C188-9 at 0, 5, or 10 uM; and RS4;11 (JAK2/CRLF2 wild-type ALL cell line). C. IC50 determination by ATP assay for C188-9 in the ALL cell lines MHH-CALL-4 and RS4;11. Each experiment was performed in triplicate. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document