Downregulation of SOX7 by DNA Methylation and Its Tumor Suppressing Role In Acute Myeloid Leukemia

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4189-4189
Author(s):  
Tsz-Kan Fung ◽  
Kin-Pong Fan ◽  
HaiXia Wan ◽  
Howard C.H. Chow ◽  
Anders S.Y. Wong ◽  
...  

Abstract Abstract 4189 Background: The SOX (Sry-related HMG box) gene family is a group of transcription factors containing in common a High-Mobility-Group (HMG) box domain which shares more than 60% homology to that in Sry. SOX proteins are involved in diverse embryonic processes and recently Sox7 was shown to regulate hematopoietic stem and progenitor cells during mouse development. In this study, we examined the expression, regulation and function of SOX7 in human acute myeloid leukemia with a view to understand its link to leukemogenesis. Method: Bone marrow (BM) or blood samples from patients with primary hematological malignancies, as well as cord blood obtained from normal Caesarian Sections were prospectively collected and mononuclear cells (MNC) fractions were obtained. Screening for SOX gene expression was performed by reverse-transcription polymer chain reaction (RT-PCR) and SOX7 expression in different experiments was further evaluated by quantitative real-time RT-PCR. Methylation of CpG islands around the sox7 transcription start site was studied by bisulphate DNA sequencing and methylation-specific PCR. Leukemic cell-lines (KG1, ML2, K562) and primary AML samples were treated with demethylating agent 5-aza-2′-deoxycytidine (5AdC). Cell proliferation of GFP or GFP-SOX7 expressing K562 cells was evaluated by SNARF-1 staining, cell-cycle analysis, 3H-thymidine incorporation and clonogenic assays. Apoptosis were evaluated by Annexin V/7-AAD assay. Canonical wnt activity of K562 cells expressing GFP or GFP-SOX7 was measured by TOP-FLASH dual luciferase assay. Result: The expression of 19 SOX genes was tested by RT-PCR in normal umbilical cord blood (UCB) as well as bone marrow or blood samples from patients with hematological malignancies. SOX7 was uniquely expressed in CD34+ cells from UCB (N=11) and most case of precursor B-cell acute lymphoblastic leukemia (ALL) (17 out of 20 tested) and a ALL derived cell line Nalm-20, but not any case of acute myeloid leukemia (N=22), myelodysplastic syndrome (N=16) or chronic myelogenous leukemia (N=13). In myeloid leukemia cell lines (KG1, ML2, K562) and primary AML samples, but not Nalm-20, the transcription start site of SOX7 contained CpG islands which were heavily methylated. Treating myeloid leukemic cell lines with 5AdC induced SOX7 expression. Enforced expression of SOX7 in K562 cells by lentiviral transduction significantly reduced cell proliferation as shown by cell growth in cultures (SOX7: 6.5-fold increase; GFP: 21.7-fold increase on Day 9, N=2), SNARF-1 staining (SOX7: 57.5%; GFP: 78.0% of total cells divided twice, N=2), 3H-thymidine incorporation assay (SOX7: 3987 cpm; GFP: 5767 cpm, N=2) and colony-forming unit (SOX7: 262±99 per 1000 input cells; GFP: 464±145 per 100 input cells, p=0.055). It also induced cell cycle delay in S/G2/M phases (SOX7: 53.4±0.35%; GFP: 44.4±2.28%, p=0.029). Apoptosis was not affected. SOX7 expression in K562 cells significantly reduced canonical-wnt activity as measured by TOP-FLASH dual luciferase assay (SOX7: 30.0±7.1-fold to FOP-FLASH; GFP: 130±18.8-fold to FOP-FLASH, p=0.0081). Conclusion: SOX7 expression in AML was regulated by promoter hypermethylation and its forced expression in K562 cells reduced cell proliferation and inhibited the canonical wnt signaling pathway. The pathogenetic link between SOX7 gene silencing and AML leukemogenesis is being investigated in our laboratory. Acknowledgments The project was supported by a grant from the strategic Research Theme of cancer stem cells in the HKU. Disclosures: No relevant conflicts of interest to declare.

2021 ◽  
Vol 11 (7) ◽  
pp. 1377-1382
Author(s):  
Lixia Cao ◽  
Jing Zhang ◽  
Huijuan Ren ◽  
Yanqiu Han

miRNA has always been a hot spot research. We assessed the effect of down-regulation of miR-23b-3p on the differentiation of acute myeloid leukemia (AML). Human AML cell line U937 was divided into blank group, NC group and miR-23b-3p low expression group (transfected with miR-23b-3p inhibitor) and miR-23b-3p followed by analysis of WT1 level and relationship between miR-23b-3p and WT1 by dual luciferase reporter assay. All-trans retinoic acid is used to induce differentiation, and then the morphological changes of cells and CD11b level were detected. When miR-23b-3p level was reduced, WT1 mRNA and protein level was also decreased. Dual luciferase assay showed that miR-23b-3p bound to WT1 3’-UTR. Inhibition of miR-23b-3p significantly decreased cell proliferation. Swiss Giemsa staining showed that most of cells were in the differentiation stage with low miR-23b-3p expression. The differentiation marker CD11b was significantly higher than other groups, indicating that low miR-23b-3p expression can promote cell differentiation and reduce cell proliferation to a certain extent. Under low miR-23b-3p expression, the positive rate of CD11b was significantly increased. Down-regulating miR-23b-3p can inhibit WT1 to a certain extent and promote the differentiation of AML, which provides a guidance for the gene-level treatment of AML.


2018 ◽  
Vol 96 (6) ◽  
pp. 840-846 ◽  
Author(s):  
Qian Ding ◽  
Qing Wang ◽  
Yi Ren ◽  
Hong Qian Zhu ◽  
ZhuYun Huang

Acute myeloid leukemia (AML) has a 5-year survival rate of only about 30%–40% due to the self-renewal and differentiation ability of leukemia stem-like cells (LSCs). To address the potential for novel therapeutic targets in LSCs, we investigated the roles of miRNA-126 and tumor necrosis factor receptor-associated factor 7 (TRAF7) in AML. We used qRT-PCR and Western blot to investigate the expression levels of miRNA-126 and TRAF7 in AML cell lines. Then, we uncovered the effect of miRNA-126 on AML cell proliferation and apoptosis by MTT assay and flow cytometric analysis, respectively. Furthermore, dual-luciferase assay and Western blot were used to determine the target of miRNA-126 in AML and the potential mechanism by which cell apoptosis is suppressed by miRNA-126. We found that miRNA-126 was highly expressed in all of the AML cell lines, and that inhibition of miRNA-126 significantly induced cell death through apoptosis. The suppression of apoptosis in AML with high expression of miRNA-126 was caused by down-regulating TRAF7, which blocked the c-FLIP pathway. The role of miRNA-126 in AML makes it a potential therapeutic target to improve clinical outcomes for patients with AML.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2951-2951
Author(s):  
Raoul Tibes ◽  
Ashish Choudhary ◽  
Amanda Henrichs ◽  
Sadia Guled ◽  
Irma Monzon ◽  
...  

Abstract In order to improve treatment strategies for Acute Myeloid Leukemia (AML), we adapted a functional genomics approach using RNAi screening to identify molecular targets that are vital to the growth of AML. Herein we report the first large-scale kinome gene silencing screen in AML. A high throughput RNAi screen was developed for the efficient siRNA transfection of AML cell lines. Eight commercially available cationic lipid-based transfection reagents were tested for their ability to transfect several AML cell lines with siRNA. These extensive transfection optimization experiments identified two AML cells lines TF-1 and ML4 with up to 95–100 and 70–75% transfection efficiency respectively. Two independent replicate kinome screens were performed on both cell lines using a siRNA library targeting 572 kinase genes with 2 siRNA/gene. At 96 hours post transfection, cell proliferation was assessed and the B-score method was used to background correct and analyze the screening data. Several siRNA to specific kinases were identified that significantly inhibit cell proliferation of up to ~40–88%. Hits were defined at two thresholds: siRNA having a B-score of <−2 providing a statistically significance of p<0.05 (confidence of > 95%) and a cutoff B-score of <−1.5 providing greater than 87% confidence for each siRNA hit. Two different kinases (2 siRNA/gene/screen) were identified as major growth regulating kinases in TF1 cells with all 4 siRNA/gene having a B-score <−2. For two additional kinases, 3/4 siRNA for each gene had a Bscore <−2. Expanding the cutoff to a B-score <−1.5 three further kinases were targeted by at least 3/4 siRNA/gene. Similar analysis using the same criteria for ML4 cells identified one kinase targeted by 3/4 siRNA at a B-score <−2, seven kinases with 2/4 siRNA <−2 and two kinases with 3/4 siRNA/gene at a B-score of <−1.5. Common hits for both cell lines with at least 6/8 siRNA per gene from 4 screens performing at a B-score <−2 identified two kinases, one of them PLK1. Applying a B-score threshold of <−1.5, we identified five kinases for which at least 5/8 siRNA/gene from 4 screens met these criteria. Kinases/genes will be presented at the meeting.Confirmation of gene silencing and validation of growth response is currently underway for a subset of genes. Among the strongest hits are siRNA targeting PLK1, as well as siRNA targeting three other kinase-genes involved in regulating cell cycle progression and checkpoints and gene ontology (GO) analysis showed enrichment in cell cycle and cell cycle-checkpoint processes. Inhibitors against PLK1 and other kinase hits identified in the screen are in (pre)-clinical development and if confirmed, our experiments provide a strong rational to test these in AML. The application of RNAi based screening is useful in the identification of genes important in AML proliferation, which could serve as targets for therapeutic intervention and guide AML drug development. Furthermore, results from these types of functional genomics approaches hold promise to be rapidly translated into clinical application.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2304-2304
Author(s):  
Amit Sharma ◽  
Nidhi Jyotsana ◽  
Courteney K. Lai ◽  
Anuhar Chaturvedi ◽  
Kerstin Görlich ◽  
...  

Abstract Introduction: Hematopoietic stem/progenitor cell differentiation is blocked in acute myeloid leukemia (AML) resulting in cytopenias and high risk of death. Most patients with AML become resistant to treatment due to lack of effective cytotoxic and differentiation fostering compounds. High expression of MN1 confers poor prognosis to AML patients and induces resistance to cytarabine and all-trans-retinoic acid (ATRA) induced differentiation. We thus set out to identify compounds which could potentially overcome the differentiation block in AML. Methods: Based on the above concepts and in an effort to identify novel compounds which are potent inducers of differentiation and apoptosis in AML, high-throughput drug screening was employed in the MN1 leukemic model. A total of 3580 bioactive compounds were tested in duplicate at a concentration of 2.5 µM using alamar blue fluorescence as readout. As MN1 cells are resistant to ATRA (at 1µM and even 10µM ATRA), the drug screening was performed in the presence of a clinically relevant dose of ATRA (1 µM) to identify compounds that concurrently act with the cytotoxic and/or differentiating effects of ATRA. To determine whether a compound was effective as monotherapy or if it synergized with ATRA, we also performed a validation phase study in which the IC50 of each candidate compound was tested alone and in combination with ATRA. Fifty-four inhibitors were chosen from the primary screen for further validation based on presumed mechanism of action and novelty. The shortlisted compound pyrimethamine (PMT) was validated for its differentiation and apoptosis promoting effects in various murine and human AML models. Results: Our high-throughput drug screening identified 117 compounds, which reduced MN1 leukemic cell proliferation by more than 80% above the ATRA-treated control in both replicates (inhibitors), 8 borderline inhibitors (one replicate with more than 80% inhibition and one with 74 to 80% inhibition), and 35 outliers, which inhibited cell proliferation by 80% or more in only one replicate. The biologic processes most frequently targeted by the 117 inhibitors were DNA replication (n=26), microtubule assembly (n=12), NF-kB pathway (n=8), dihydrofolate reductase (DHFR, n=3) and heat shock protein 90 (HSP90). Dihydrofolate reductase inhibitors, pyrimethamine and amethopterin/methotrexate emerged as top hits from the screening and preliminary validation studies. Validation studies identified the antifolate pyrimethamine (PMT) that potently induced apoptosis and differentiation in several murine and human leukemic cell lines when administered as a single agent. The cytotoxic effects of pyrimethamine were reversed by addition of an excess of folic acid whereas induction of myeloid differentiation at higher concentrations of pyrimethamine was not mediated through DHFR inhibition. We further evaluated the effect of pyrimethamine in an in vivo xenograft mouse model by subcutaneously inducing tumors with HL60 and THP1 cell lines. Oral pyrimethamine treatment significantly reduced tumor volumes after 14, 19 and 24 days post-transplantation and at death compared to solvent treated mice (P<0.01). The effect of pyrimethamine was further assessed in primary human AML cells and normal CD34+ cells by CFC assays. Colony numbers from primary AML cells, but not normal CD34+ bone marrow cells, were significantly reduced by pyrimethamine as compared to solvent control. Thus, our study identifies pyrimethamine as a candidate drug that is a potent and specific inducer of apoptosis and differentiation with the property of specifically targeting leukemic cells. Conclusion: Our high-throughput drug screening identified pyrimethamine as a potent and specific antileukemic compound and reinforces targeting of folate metabolism as a treatment strategy in acute myeloid leukemia. Disclosures No relevant conflicts of interest to declare.


Author(s):  
Xiaomin Chen ◽  
Fengqi Liu ◽  
Dade Rong ◽  
Limei Xu ◽  
Xiuzhen Tong ◽  
...  

Background: SH3-domain-binding glutamic acid-rich protein-like protein (SH3BGRL) is downregulated in acute myeloid leukemia (AML). Clinically, DNA demethylating drug decitabine (DAC) combined with traditional chemotherapies reveals better efficacy on AML patients than the conventional chemotherapies alone. Our previous results revealed that human SH3-domain-binding glutamic acid-rich protein-like protein (SH3BGRL) plays a tumor suppressive role in AML but whether there is a connection between DAC and SH3BGRL expression remains elusive. Methods: Here, we tentatively treated AML cell lines U937, MV4, and HL-60 with DAC and Western Blots. RT-PCR was used to detect the expression of SH3BGRL. Cell proliferation and apoptosis were determined using Annexin V/7-AAD staining. Real-time RT-PCR and Western blot were used to determine the expression of SH3BGRL mRNA and protein. Methylation-specific PCR was used to quantify the DNA methylation in AML cell lines.Results: DAC had cytotoxicity in HL-60, MV4, and U937. In U937 cell lines, treatment with DAC showed the up-regulation of caspase, PARP, and SH3BGRL. Upon treatment, up-regulation of SH3BGRL mRNA and protein was dose-dependent and this activity was partially inhibited in endogenous SH3BGRL knockdown cell lines. Results: DAC had cytotoxicity in HL-60, MV4, and U937. In U937 cell lines, treatment with DAC showed the up-regulation of caspase, PARP, and SH3BGRL. Upon treatment, up-regulation of SH3BGRL mRNA and protein was dose-dependent and this activity was partially inhibited in endogenous SH3BGRL knockdown cell lines. Conclusion: Thus, our results demonstrated a possibly cytotoxic role of DAC on AML cells by upregulation of SH3BGRL expression at epigenetic modulation level and the methylation status in the SH3BGRL promoter region could be a supplemental diagnostic marker to the precise administration of DAC to AML patients.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3688-3688
Author(s):  
Jonathan L. Metts ◽  
Heath L. Bradley ◽  
Neil P. Shah ◽  
Reuben Kapur ◽  
Jack L. Arbiser ◽  
...  

Abstract Acute myeloid leukemia (AML) is the most common acute leukemia in adults and the 2nd most common in children. Despite advances in our understanding of AML biology, long term survival remains suboptimal. Current therapy primarily involves high dose cytotoxic chemotherapy and possible allogeneic stem cell transplant, both of which are heavily myelosuppressive and carry some treatment related mortality. The FLT3/ITD mutation is found in 20-30% of adults and 10-20% of children with AML, and despite intensive therapy survival outcomes are dismal. Recent strategies using targeted therapies (e.g. FLT3 tyrosine kinase inhibitors-TKIs) have so far yielded modest responses, and most relapsed/refractory patients will still die of their leukemia. Thus selective agents targeting FLT3 mutated AML but not benign hematopoiesis are needed. A large portion of AML, including those with FLT3/ITD, has elevated levels of reactive oxygen species (ROS), and AML with high ROS is highly proliferative. Interestingly, mislocalization of mutant type III receptor tyrosine kinases (RTKs, e.g. FLT3 and c-KIT) occurs in leukemia. In FLT3/ITD, a portion of underglycosylated RTK accumulates on the endoplasmic reticulum (ER) in a microenvironment with high levels of localized ROS generated from ER-bound NADPH Oxidase 4 (Nox4) and through tight connections with mitochondria. ROS has been demonstrated in recent years to be an integral part of cell signaling, typically through suppression of protein tyrosine phosphatases. Therefore, we hypothesized that elevated ROS plays an important role in leukemic cell signaling in FLT3/ITD+ AML. To this end, we have investigated the effects of the Nox4 inhibitor imipramine blue (IB), a novel potent agent from the class of triphenylmethane dyes (Munson et al, Sci Transl Med, Vol. 4, 2012). We used FLT3/ITD+ AML cell lines (MV4-11, MOLM-14), cell lines without FLT3/ITD (OCI-AML3, K562, HEL, HL-60), and Ba/F3 and 32D cells transduced with FLT3 and c-KIT mutants, respectively. We screened inhibitors of mitochondrial complex I and NOX inhibitors to determine their effects on ROS levels by H2-DCF-DA staining/flow cytometry and on cell viability by trypan blue exclusion assay. While all mitochondrial and NOX4 inhibitors tested effectively reduced total cellular ROS levels, only Nox4 inhibition with IB achieved selective cytotoxicity for FLT3/ITD+ cell lines with high potency (IC50 =125 nM in MV4-11). The Annexin V assay showed that IB causes cell death through an apoptotic mechanism in MV4-11 cells. To determine effects of Nox4 inhibition on leukemic cell signaling, specifically on protein tyrosine phosphatases (PTPs), we studied the signal transducer and activator of transcription 5 (STAT5). We observed phosphorylated STAT5 (pSTAT5) levels by Western blot, and sodium orthovanadate (Na3VO4) was used for PTP inhibition. IB caused dose dependent decreases in pSTAT5 levels after 4 hours of exposure, and this effect was reversed when Na3VO4 was added, implicating reactivated PTPs as the cause of pSTAT5 suppression. To determine the downstream effects of STAT5 inhibition by IB, STAT5 target genes were evaluated by qRT-PCR. Four hour IB treatment caused decreased expression of common STAT5 target genes (Pim1, c-Myc, Cish) indicating effective suppression of STAT5 function. To optimize cytotoxicity, we then combined IB with the STAT5 inhibitor pimozide. A combination of 75 nM IB/5µM pimozide was highly synergistic in MV4-11 (5 fold higher cytotoxicity over individual drugs, 3 fold higher than their additive effect). IB/pimozide selectively killed FLT3/ITD+ AML cell lines while sparing AML cells without FLT3/ITD and benign CD34+ cord blood cells. IB and IB/pimozide were also equally effective in killing Ba/F3 FLT3/ITD cells with point mutations causing resistance to TKIs and selectively killed 32D cells with c-KIT (D814V) mutation over those with wild type c-KIT. Thus we have shown that the Nox4 inhibitor IB is a potent and selective inducer of apoptosis for FLT3/ITD+ AML in vitro, and the novel combination of IB and pimozide selectively targets FLT3/ITD and c-KIT positive mutants, including TKI-resistant FLT3/ITD mutants. We propose that IB alone or in combination with pimozide represents a novel therapeutic strategy, and testing in primary patient samples and in vivo models are currently underway. Disclosures Shah: Plexxikon Inc.: Research Funding; Pfizer: Research Funding; Bristol-Myers Squibb: Research Funding. Arbiser:ABBY Therapeutics: Other: Jack L Arbiser is listed as inventor on a US Patent for imipramine blue. He is cofounder of ABBY Therapeutics, which has licensed imipramine blue from Emory University.


Blood ◽  
2009 ◽  
Vol 114 (3) ◽  
pp. 659-662 ◽  
Author(s):  
Annelies G. Renner ◽  
Cédric Dos Santos ◽  
Christian Recher ◽  
Christian Bailly ◽  
Laurent Créancier ◽  
...  

Abstract Polo-like kinase 1 (Plk1) is a major mitotic regulator overexpressed in many solid tumors. Its role in hematopoietic malignancies is still poorly characterized. In this study, we demonstrate that Plk1 is highly expressed in leukemic cell lines, and overexpressed in a majority of samples from patients with acute myeloid leukemia compared with normal progenitors. A pharmacologic inhibitor, BI2536, blocks proliferation in established cell lines, and dramatically inhibits the clonogenic potential of leukemic cells from patients. Plk1 knockdown by small interfering RNA also blocked proliferation of leukemic cell lines and the clonogenic potential of primary cells from patients. Interestingly, normal primary hematopoietic progenitors are less sensitive to Plk1 inhibition than leukemic cells, whose proliferation is dramatically decreased by the inhibitor. These results highlight Plk1 as a potentially interesting therapeutic target for the treatment of acute myeloid leukemia.


Author(s):  
Mat Jusoh Siti Asmaa ◽  
Hamid Ali Al-Jamal ◽  
Abdul Rahim Hussein ◽  
Badrul Hisham Yahaya ◽  
Azlan Husin ◽  
...  

Background: Acute myeloid leukemia (AML) is the most common form of acute leukemias in adults which is clinically and molecularly heterogeneous. Several risk and genetic factors have been widely investigated to characterize AML. However, the concomitant epigenetic factors in controlling the gene expression lead to AML transformation was not fully understood. This study was aimed to identify epigenetically regulated genes in AML cell lines induced by epigenetic modulating agents, Trichostatin A (TSA) and 5-Azacytidine (5-Aza). Materials and Methods: MV4-11 and Kasumi 1 were treated with TSA and/or 5-Aza at IC50 concentration. Gene expression profiling by microarray was utilized using SurePrint G3 Human Gene Expression v3. Gene ontology and KEGG pathway annotations were analyzed by DAVID bioinformatics software using EASE enrichment score. mRNA expression of the differentially expressed genes were verified by quantitative real time PCR. Results: Gene expression analysis revealed a significant changes in the expression of 24,822, 15,720, 15,654 genes in MV4-11 and 12,598, 8828, 18,026 genes in Kasumi 1, in response to TSA, 5-Aza and combination treatments, respectively, compared to non-treated (p<0.05). 7 genes (SOCS3, TUBA1C, CCNA1, MAP3K6, PTPRC, STAT6 and RUNX1) and 4 genes (ANGPTL4, TUBB2A, ADAM12 and PTPN6) shown to be predominantly expressed in MV4-11 and Kasumi 1, respectively (EASE<0.1). The analysis also revealed phagosome pathway commonly activated in both cell lines. Conclusion: Our data showed a distinct optimal biological characteristic and pathway in different types of leukemic cell lines. These finding may help in the identification of cell-specific epigenetic biomarker in the pathogenesis of AML.  


Sign in / Sign up

Export Citation Format

Share Document