human leukemic cell line
Recently Published Documents


TOTAL DOCUMENTS

91
(FIVE YEARS 0)

H-INDEX

23
(FIVE YEARS 0)

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1529-1529
Author(s):  
Zhijie Wu ◽  
Xin Zhao ◽  
Lauren G. Banaszak ◽  
Keyvan Keyvanfar ◽  
Sachiko Kajigaya ◽  
...  

Abstract Introduction .Mutations in the Additional Sex Combs Like1 (ASXL1) gene are frequent in myeloid malignances (myelodysplastic syndromes [MDS], myeloproliferative neoplasms, chronic myelomonocytic leukemia and acute myeloid leukemia [AML]), and they predict poor survival. As we recently published (Yoshizato T, et al., N Engl J Med 2015), mutations in ASXL1 as well as BCOR/BCORL1, PIGA, and DNMT3A are most prevalent in patients with aplastic anemia (AA). ASXL1 grouped with "unfavorable mutations" that conferred poor survival and increased risk of "clonal evolution" of AA to MDS and AML. As an epigenetic modifier, ASXL1mutations may be involved in myeloid malignant transformation (Davies C, et al., Br J Haematol 2013; Abdel-Wahab O, et al., Cancer Cell 2012) but precise mechanisms have not been delineated. Methods.The CRISPR/Cas9 system was employed to generate ASXL1-knockout clones from the U937 human leukemic cell line. Single cell clones were sorted by flow cytometry, and ASXL1 mutations were assessed by Sanger sequencing. Clones with mutations that encoded truncated proteins were used for further experiments. Characteristic features of both mutated and wild-type (WT) clones were examined: cell morphology by Wright-Giemsa staining; karyotype analysis by G-banding; and cell cycle, apoptosis, and cell differentiation by flow cytometry. RNA sequencing (RNA-Seq) was performed to screen differentially expressed genes between WT and ASXL1-knockout clones, followed by validation of gene expression using reverse transcription quantitative PCR (RT-qPCR). Results.Of 88 single cell clones, 23 clones exhibited frame-shift and nonsense mutations of ASXL1, resulting in truncated proteins. Among them, 17 clones with single nucleotide insertion (c.594insA, heterozygous or homozygous) that encoded truncated proteins Ser199Glufsx55 were used for further experiments. Karyotype analysis revealed no significant differences between WT and ASXL1-knockout clones, suggesting that neither CRISPR/Cas9 itself nor ASXL1 mutations caused chromosomal instability. No significant differences between WT and ASXL1-knockout clones were observed in cell morphology, cell proliferation, cell cycle, or 5-fluoruracil-induced cell apoptosis. When monocyte/macrophage differentiation was induced chemically by exposure to phorbol 12-myristate 13-acetate, CD11b cell surface expression was much lower in ASXL1-knockout clones than in WT. By RNA-Seq, several genes (BIRC7, CACNA2D3, CTSG, CYBB, NAIP, NTNG1, OXR1, and ACTL8)involved in pathways related to cell death and survival (including TNFR1 signaling pathway, TNFR2 signaling pathway, TWEAK signaling pathway, apoptosis signaling pathway, death receptor signaling pathway, Rac signaling pathway, and phagosome maturation pathway) were down-regulated, but were not correlated to functional deficits in cell growth or apoptosis, which was indistinguishable between ASXL1-knockout and WT clones. RT-qPCR confirmed down-regulation of genes in ASXL1-knockout compared to WT clones, including CYBB (restrictedly expressed in terminally differentiating myeloid cells) and CLEC5A (involved in granulocytic differentiation), which may contribute to disturbance in myeloid differentiation of ASXL1-knockout U937 cells. Downregulaton of NAIP, which inhibits CASP3, CASP7, and CASP9 activities, may be related to deregulation of apoptosis in ASXL1-knockout U937. Conclusion. ASXL1 mutations are frequent in both malignant and non-malignant blood diseases, and may be involved in myeloid neoplastic transformation. Using gene editing of a human leukemic cell line, we show that ASXL1-knockout perturbs myeloid differentiation and down regulates multiple genes associated with myeloid development such as CYBB and CLEC5A, providing potential mechanisms for its role in bone marrow failure and neoplastic transformation. Disclosures Young: Novartis: Research Funding.


2015 ◽  
Vol 29 (7) ◽  
pp. 1545-1554 ◽  
Author(s):  
Andrea Fernández-Araujo ◽  
Amparo Alfonso ◽  
Mercedes R. Vieytes ◽  
Luis M. Botana

ISRN Oncology ◽  
2013 ◽  
Vol 2013 ◽  
pp. 1-5
Author(s):  
Mark R. Garbrecht ◽  
Thomas J. Schmidt

Glucocorticoids are commonly used in the first-line treatment of hematological malignancies, such as acute lymphoblastic leukemia, due to the ability of these steroids to activate pro-apoptotic pathways in human lymphocytes. The goal of the current study was to examine the gene expression and enzyme activity of the microsomal enzyme, 11-β hydroxysteroid dehydrogenase type 2 (HSD11B2, HSD2), which is responsible for the oxidation of bioactive glucocorticoids to their inert metabolites. Using the glucocorticoid-sensitive human leukemic cell line, CEM-C7, we were able to detect the expression of HSD2 at the level of mRNA (via RT-PCR), protein (via immunohistochemistry and immunoblotting), and enzyme activity (via conversion of tritiated cortisol to cortisone). Furthermore, we observed that HSD2 enzyme activity is down regulated in CEM-C7 cells that were pretreated with the synthetic glucocorticoid, dexamethasone (100 nM, <15 hours), and that this down regulation of enzyme activity is blocked by the administration of the glucocorticoid receptor antagonist, RU-486. Taken collectively, these data raise the possibility that the effectiveness of glucocorticoids in the treatment of human leukemias may be influenced by: (1) the ability of these neoplastic cells to metabolize glucocorticoids via HSD2 and (2) the ability of these steroids to regulate the expression of this key enzyme.


2012 ◽  
Vol 237 (8) ◽  
pp. 933-942 ◽  
Author(s):  
Pilar Sancho ◽  
Eva Galeano ◽  
María Cristina Estañ ◽  
Irene Gañán-Gómez ◽  
María del Carmen Boyano-Adánez ◽  
...  

2012 ◽  
Vol 2012 ◽  
pp. 1-11 ◽  
Author(s):  
Qing Xia ◽  
Zhi-Yong Wang ◽  
Hui-Qing Li ◽  
Yu-Tao Diao ◽  
Xiao-Li Li ◽  
...  

Multidrug resistance (MDR) is the major obstacle in chemotherapy, which involves multiple signaling pathways. Diallyl trisulfide (DATS) is the main sulfuric compound in garlic. In the present study, we aimed to explore whether DATS could overcome P-glycoprotein-(P-gp-)mediated MDR in K562/A02 cells, and to investigate whether NF-κB suppression is involved in DATS-induced reversal of MDR. MTT assay revealed that cotreatment with DATS increased the response of K562/A02 cells to adriamycin (the resistance reversal fold was 3.79) without toxic side effects. DATS could enhance the intracellular concentration of adriamycin by inhibiting the function and expression of P-gp, as shown by flow cytometry, RT-PCR, and western blot. In addition, DATS resulted in more K562/A02 cell apoptosis, accompanied by increased expression of caspase-3. The expression of NF-κB/p65 (downregulation) was significantly linked to the drug-resistance mechanism of DATS, whereas the expression of IκBαwas not affected by DATS. Our findings demonstrated that DATS can serve as a novel, nontoxic modulator of MDR, and can reverse the MDR of K562/A02 cells in vitro by increasing intracellular adriamycin concentration and inducing apoptosis. More importantly, we proved for the first time that the suppression of NF-κB possibly involves the molecular mechanism in the course of reversion by DATS.


2010 ◽  
Vol 34 (8) ◽  
pp. 1052-1056 ◽  
Author(s):  
Roula M. Abdel-Massih ◽  
Rida Fares ◽  
Samer Bazzi ◽  
Nisrine El-Chami ◽  
Elias Baydoun

Sign in / Sign up

Export Citation Format

Share Document