Skip to main content
International Journal of Hematology-Oncology and Stem Cell Research logoLink to International Journal of Hematology-Oncology and Stem Cell Research
. 2020 Jan 1;14(1):72–92.

Transcriptomic Profiles of MV4-11 and Kasumi 1 Acute Myeloid Leukemia Cell Lines Modulated by Epigenetic Modifiers Trichostatin A and 5-Azacytidine

Mat Jusoh Siti Asmaa 2,3,4, Hamid Ali Al-Jamal 2,3,4, Abdul Rahim Hussein 2,3,4, Badrul Hisham Yahaya 2,3,4, Roslin Hassan 1, Faezahtul Arbaeyah Hussain 5, Shaharum Shamsuddin 6,7, Muhammad Farid Johan 1
PMCID: PMC7167603  PMID: 32337016

Abstract

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.

Key Words: Acute myeloid leukemia, Epigenetics* Histone deacetylase inhibitors, 5-Azacytidine, Gene expression

Introduction

Acute myeloid leukemia (AML) is characterized by a block in early progenitor differentiation leading to accumulation of immature and highly proliferative leukemic stem cells (LSCs) in the bone marrow and peripheral blood   1 . The 2017 World Health Organization (WHO) has provided guidelines on the cut-off value of blast percentage of AML by; 200 and 500 cells-leukocytes differential counts in the peripheral blood and in the bone marrow, respectively   2 . For a diagnosis of AML, a marrow or blood blast count of 20% or more is required, except for AML with t(15;17), t(8;21), inv(16) or t(16;16), and some cases of erythroleukemia. AML is the most common form of acute leukemias in adults which affected 32% adults. Although the overall mortality rate has decreased by 1.0% each year from 2001 to 2010, the overall incidence rate was increased by 0.2% each year. In 2018, the American Cancer Society estimated that 19,520 of new cases and 10,670 deaths from AML. The 5-years overall survival rate was also poor with only 24%   3 .

For many years, gene expression profiling by microarray was used as a traditional method to search abnormalities in cancers, including in AML   4 . These presented data was invaluable and accessible to the identification of disease’s class discovery, class prediction, and class comparison. Class discovery refers to the identification of a new subgroup, that later was class predicted by gene expression data. The first and second class already had a diagnostic implication. While the third class, which is class comparison refer to the identification of genes that were deregulated in certain subgroups, that may address biological function   5 .

It has long established that AML is clinically heterogeneous disease characterized by an accumulation of continuous genetic abnormalities   6  and prior epigenetic lesions   7  resulting in clonal evolution and expansion. The considerable complexities disrupt the genetic and epigenetic landscapes by changes in gene expression   8  which profoundly affecting treatment response and patients’ survival. Earlier epigenetic alteration established cellular identities initiating tumorigenesis by inappropriate activation or inhibition of cellular signaling pathways   9 . For example, promoter hypermethylation of a tumor suppressor genes is commonly implicated in cancer   10 , involving genes controlling the cell cycle and DNA repair   11 . On the other hand, modification to histone protein in nucleosome modulates the transcriptional burst frequency specifically through histone acetylation   12 . Both epigenetic mechanisms endow the regulation in gene expression. Hence, targeting the epigenetically-regulated genes in the control of AML licensed a promising outcome.

In this study, high-throughput microarray technique was used to analyze epigenetic-derived molecular mechanism by modulating gene expression using a classical DNA methyltransferase (DNMT) inhibitor; 5-Azacytidine (5-Aza) and a histone deacetylase (HDAC) inhibitor, Trichostatin A (TSA). The aim of this study was to induce the epigenetic response via gene re-expression or down-expression in two types of AML cell lines; MV4-11 and Kasumi 1. It was hypothesized that the silencing of a tumor suppressor gene and the activation of oncogenes in AML were due to epigenetic mechanisms of DNA hypermethylation and histone deacetylation.

MATERIALS AND METHODS

MV4-11 and Kasumi 1 cell culture

MV4-11 is a human AML cell line established from blasts cells of 10 years old male with biphenotypic B-myelomonocytic leukemia (AML FAB M5) that carry translocation t(4;11) and a FLT3-ITD mutation. Kasumi 1 is a human AML cell line established from peripheral blast cells from 7 years old juvenile male Japanese that carry translocation t(8;21) and AML1-ETO (also known as RUNX1-CBF2T1) fusion genes. The AML cell lines were originally purchased from the American Type Culture Collection (ATCC, VA, USA). Both AML cell lines were cultured in RPMI-1640 (Gibco®, CA, USA) supplemented with 10% Fetal bovine serum (Sigma-Aldrich, MO, USA) and 0.1% penicillin/streptomycin (Invitrogen, CA, USA) in humidified temperature containing 5% carbon dioxide (CO2) at 37°C.

TSA and/or 5-Aza treatment

TSA (Sigma-Aldrich, MO, USA) and 5-Aza (Sigma-Aldrich, MO, USA) were dissolved in DMSO (Sigma-Aldrich, MO, USA) and RPMI-1640, respectively to a stock concentration of 500 µM, and further diluted to the desired working concentrations. MV4-11 and Kasumi 1 were seeded in 6-wells plate to 80-90% confluency at the initial cell number of 1 x 105 cells/mL prior to the drug treatment for 24 hours. The cell lines were treated with varying concentration of TSA (0, 1.25, 2.5, 5.0, 10.0 µM) and 5-Aza (0, 5.0, 10.0, 20.0, 50, 100 µM) and incubated for 24 hours under humidified temperature.

Cell Viability Assay

Percentage viability of non-treated and treated MV4-11 and Kasumi 1 after the 24 hours exposure to TSA and 5-Aza treatments were measured by Trypan Blue Exclusion Assay (Life Technologies, CA, USA). The half maximal inhibitory concentration (IC50) was determined by GraphPad Prism 6.0 (GraphPad, CA, USA).

Total RNA extraction and quality control

Total RNA was extracted from treated and untreated MV4-11 and Kasumi 1 using Total RNA Isolation Kit (Promega, SA, USA) according to the manufacturer’s protocol. The final elution step was performed using 30 µl of elution buffer for a highly concentrated RNAs. The isolated RNA concentration and purity were determined by Nanodrop ND-1000 spectrophotometer (Thermo-Fisher Scientific, WA, USA). Prior to the gene expression profiling, the RNA integrity was assessed by 1.5% agarose gel electrophoresis and their RIN (RNA integrity number) values were determine by Agilent 2100 Bioanalyzer (Agilent, CA, USA). The qualified RNAs (absorbance 280/260 1.8-2.1 ratio; highly intact 28S and 18S ribosomal RNA and RIN above 7) were stored at -80 ºC until further analysis.

Microarray analysis

Whole genome expression profiling was performed using One-Color SurePrint G3 Human Gene Expression v3, 8 x 60K slides contained array probe (Agilent Technologies, CA, USA). Prior to Cyanine 3 (Cy3) labeling, RNA spiked-In dilution was prepared using RNA spiked-In Kit (Agilent Technologies, CA, USA) to each sample using T7 RNA polymerase (RNA reference target) for normalization. Cy3-labeled cRNA was generated from 25 ng input total RNA using Low Input Quick Amp Labeling Kit (Agilent Technologies, CA, USA). The fluorescent-labeled cRNA was purified by RNAeasy Mini Kit and RNAase-free DNAase Set (Qiagen, CA, USA) and quantified by Nanodrop ND-1000 spectrophotometer. 25 ng of fluorescein-labeled and amplified cRNA was hybridized into array slides containing 60,000 probes (Agilent Technologies, CA, USA) at 65 degree Celsius for 17 hours. After hybridization and washing steps, the array slides were scanned using SureCan Microarray Scanner (Agilent Technologies, CA, USA) to measure the fluorescence intensity of Cy3 labeled RNA bound to the microarray slide. The resulted images were processed using the Feature Extraction (FE) software v.12 (Agilent Technologies, CA, USA) for data filtering. Raw data obtained was analyzed by Genespring GX v12.6 software (Agilent Technologies, CA, USA).

Database screening

Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis annotations were utilized by the Database for Annotation, Visualization and Integrated Discovery (DAVID) Bioinformatics Resources v6.8 (https://david.ncifcrf.gov/) to characterize and predict epigenetically regulated genes in treated AML cell lines. The Enhanced AL Scoring Engine (EASE) scoring system (a modified Fisher Exact p-value, p<0.1) was implemented for statistical analysis to provide enriched GO terms and pathways annotation within gene lists. EASE analysis produces a consistent and similar functional annotation with numerous analytical methods   13 , and Venn diagram was constructed to analyze genes with differential expression pattern after TSA and 5-Aza treatment in MV4-11 and Kasumi 1. The analysis was conducted by the Venny 2.1 software (http://bioinfogp.cnb.csic.es/tools/venny/).

Quantitative Real-time PCR (qRT-PCR)

To validate microarray data, qRT-PCR analysis on selected up-regulated and down-regulated genes was performed by Taqman gene expression assays and analyzed using Applied Biosystem (ABI)® 7500 Real-Time PCR Machine (Applied Biosystem, CA, USA). Total RNAs from untreated and treated cell lines were reverse transcribed using High-Capacity cDNA Reverse Transcription Kit (Applied Biosystem, CA, USA). Pre-designed assays (PrimeTime® Pre-designed Assays) (IDT Inc., IA, USA) [ANGPTL4 (assay ID: Hs.PT58.25480012), TUBB2A (assay ID: Hs.PT58.40767003), PTPN6 (assay ID: Hs.PT58.23073507) and ADAM12 (assay ID: Hs.PT58.26423628)], and custom-designed primers and probes (SOCS3, TUBA1C, CCNA1, MAP3K6, STAT6, PTPRC and RUNX1 genes) were amplified by PrimeTime® Gene Expression Master Mix (IDT Inc., IA, USA). Assay sequences were confirmed using web Basic Local Alignment Search Tool (BLAST) by the National Center for Biotechnology Information (NCBI) (U.S. National Library of Medicine, MD, USA). The qRT-PCR amplification conditions were: 95°C for 3 min for enzyme activation, 40 cycles of denaturation at 95°C for 15 s and 60°C for 1 min for annealing and extension. B2M and GAPDH were used as endogenous control genes and expression levels were estimated using relative quantitation (RQ) of duplicated samples calculated by 2-∆∆CT method (∆∆CT=∆CTTreated–∆CTUntreated, ∆CT=CtSelected Genes –CtB2M/GAPDH).

Results

A significant decrease in cell viability was observed after the TSA and 5-Aza treatments (One-way ANOVA, p<0.05). The half maximal inhibitory concentration (IC50) was acquired at 2.2 µM and 2.3 µM for MV4-11 and; 6.25 µM and 6.95 µM for Kasumi 1 in TSA and 5-Aza, respectively. TSA and 5-Aza treatments have higher potency in MV4-11 due to their lower IC50 value compared to Kasumi 1 (Figure 1).

Figure 1.

Figure 1

Effect of TSA and 5-Aza treatment on cell viability by percentage (%) inhibition of MV4-11 and Kasumi 1 cell lines relative to non-treated cell lines. Significant inhibition of MV4-11 after (a) TSA and (b) 5-Aza treatment at increasing concentration (0.0, 1.25, 2.5, 5.0 and 10.0 µM) for 24 h. Significant inhibition of Kasumi 1 after (c) TSA treatment at increasing concentration (0.0, 1.25, 2.5, 5.0 and 10.0 µM) and (d) 5-Aza (0.0, 5.0, 10.0, 20.0, 50.0 and 100.0 µM) for 24 h calculated by Trypan Blue Exclusion Assay (TBEA) (One-Way ANOVA, LSD multiple comparison, p<0.05).

Gene expression profile of MV4-11 and Kasumi 1 in response to TSA and 5-Aza

The gene expression profile of MV4-11 and Kasumi 1 after 24 hours of TSA, 5-Aza and combination (TSA+5-Aza) treatments at IC50 concentration. The exploratory microarray analysis was carried out to short-list the differentially expressed genes induced by the drug treatments analyzed by GeneSpring software 12.1 (the cut-off value; fold change ≥ 2.0, significance level, Pearson, P <0.05). 33,150 and 24,668 genes passed the FE filtering in MV4-11 and Kasumi 1, respectively. In MV4-11, 24,822 genes’ expressions were altered (either up or down-regulated) in TSA, 15,720 in 5-Aza and 15,654 in TSA+5-Aza. Whereas in Kasumi 1, 12,598 genes were altered in TSA, 8828 genes in 5-Aza and 18,026 genes in TSA+5-Aza treatments, normalized to non-treated cells (Figure 2). The most up-regulated and down-regulated genes in TSA, 5-Aza and TSA+5-Aza treatments and their folds change were listed in Tables 1 and 2. Genes were selected according to these three criteria: 1. Relevant genes with the highest fold-change different and commonly regulated across all treatments, 2. Relevant genes reported having an association with AML and other myeloid neoplasms from the previous study and/or Pubmed literature, 3. Genes with not otherwise classified under both criteria but could be interesting due to their implication in pathways in cancer.

Figure 2.

Figure 2

Microarray gene expression analysis for MV4-11 and Kasumi 1 treated with TSA, 5-Aza and TSA+5-Aza. Number of up-regulated and down-regulated genes was created by Genespring software analysis. Further analysis to obtain gene entities were performed using Moderated T-test with multiple correction (Benjamini Hochberg FDR) with p-value <0.05 and fold change of >2.0 as a significant.

Table 1(a).

Most up- and down-regulated genes in TSA treated MV4-11

Gene Bank
Accession
Gene symbol Gene description ( Homo sapiens) *Folds
Change
NM_001082 CYP4F2 Cytochrome P450, family 4, subfamily F, polypeptide 2 1094.05
NM_014971 EFR3B EFR3 homolog B (S. cerevisiae) 360.59
NM_006569 CGREF1 Cell growth regulator with EF-hand domain 1 348.85
NM_017702 DEF8 Differentially expressed in FDCP 8 325.92
NM_003914 CCNA1 Cyclin A1 298.44
NM_003255 TIMP2 TIMP metallopeptidase inhibitor 2 281.56
NM_031313 ALPPL2 Alkaline phosphatase, placental-like 2 250.36
NM_032704 TUBA1C Tubulin, alpha 1c 234.14
NM_003955 SOCS3 Suppressor of cytokine signaling 3 176.76
NM_001204054 NDUFC2 NADH dehydrogenase (ubiquinone) 1, subcomplex unknown 2 166.94
NR_027028 GUSBP1 Glucuronidase, beta pseudogene 1 153.18
NM_004522 KIF5C Kinesin family member 5C 153.59
NM_003520 HIST1H2BN Histone cluster 1, H2bn 150.13
NM_006321 ARIH2 Ariadne RBR E3 ubiquitin protein ligase 2 133.61
NM_000612 IGF2 Insulin-like growth factor 2 131.09
NM_177424 STX12 Syntaxin 12 103.73
NM_006086 TUBB3 Tubulin, beta 3 class III 80.38
NM_004672 MAP3K6 Mitogen-activated protein kinase kinase kinase 6 39.50
NM_001025300 RAB12 member RAS oncogene family 38.83
NM_139314 ANGPTL4 Angiopoietin-like 4 26.79
NM_018437 HEMGN Hemogen -518.75
NM_024913 CPED1 Cadherin-like and PC-esterase domain containing 1 -243.96
NM_003152 STAT5A Signal transducer and activator of transcription 5A -159.83
NM_002838 PTPRC Protein tyrosine phosphatase, receptor type C -138.75
NM_080612 GAB3 GRB2-associated binding protein 3 -117.26
NM_003126 SPTA1 Spectrin, alpha, erythrocytic 1 -107.30
NM_015401 HDAC7 Histone deacetylase 7 -88.16
NM_006563 KLF1 Kruppel-like factor 1 (erythroid) -85.08
NM_015660 GIMAP2 GTPase, IMAP family member 2 -73.83
NM_006163 NFE2 Nuclear factor, erythroid 2 -69.24
NM_213674 TPM2 Tropomyosin 2 (beta) -57.76
NM_006287 TFPI Tissue factor pathway inhibitor -55.30
NM_005021 ENPP3 pyrophosphatase/phosphodiesterase 3 -49.49
NM_004688 NMI N-myc (and STAT) interactor -47.85
NM_000037 ANK1 Ankyrin 1, erythrocytic, transcript variant 3 -46.78
NM_013427 ARHGAP6 Rho GTPase activating protein 6 -42.54
NM_006546 IGF2BP1 Insulin-like growth factor 2 mRNA binding protein 1 -42.54
NM_033306 CASP4 Caspase 4, apoptosis-related cysteine peptidase -42.42
NM_080588 PTPN7 Protein tyrosine phosphatase, non-receptor type 7 -39.69
NM_004753 DHRS3 dehydrogenase/reductase (SDR family) member 3 -36.59
NR_026812 RUNX1-IT1 RUNX1 intronic transcript 1 -22.05
NM_003153 STAT6 signal transducer and activator of transcription 6 -10.04

*Folds-change of treatment group compared to control analyzed by Genespring software analysis, Moderated T-test, p<0.05)

Table 1(b).

Most up- and down-regulated genes in 5-Aza treated MV4-11

Gene Bank Accession Gene symbol Gene description ( Homo sapiens) *Folds
change
NM_001145191 FAM200B family with sequence similarity 200, member B 461.79
NM_032905 RBM17 RNA binding motif protein 17 336.98
NM_017702 DEF8 differentially expressed in FDCP 8 homolog 277.69
NM_024097 C1orf50 chromosome 1 open reading frame 50 207.14
NM_001204054 NDUFC2 NADH dehydrogenase 185.92
NM_006321 ARIH2 ariadne RBR E3 ubiquitin protein ligase 2 158.81
NR_027028 GUSBP1 glucuronidase, beta pseudogene 1, non-coding RNA 157.88
NM_032704 TUBA1C tubulin, alpha 1c 154.28
NM_031925 TMEM120A transmembrane protein 120A 135.01
NM_003955 SOCS3 suppressor of cytokine signaling 3 120.31
NM_015046 SETX Homo sapiens senataxin 95.04
NM_016256] NAGPA N-acetylglucosamine-1-phosphodiester alpha-N-acetylglucosaminidase 93.98
NM_001031713 MCUR1 mitochondrial calcium uniporter regulator 1 92.49
NM_033028 BBS4 Bardet-Biedl syndrome 4 90.09
NM_177424 STX12 syntaxin 12 89.59
NM_003520 HIST1H2BN histone cluster 1, H2bn 89.53
NM_052936] ATG4A autophagy related 4A, cysteine peptidase 85.61
NM_014884 SUGP2 SURP and G patch domain containing 2 70.67
NM_138501 TECR trans-2,3-enoyl-CoA reductase 69.28
NM_004672 MAP3K6 mitogen-activated protein kinase kinase kinase 6 48.45
NM_005614 RHEB Homo sapiens Ras homolog enriched in brain 45.97
NM_013230 CD24 CD24 molecule 45.50
NM_001025300 RAB12 RAB12, member RAS oncogene family 44.06
NM_173698 FAM133A family with sequence similarity 133, member A -101.93
NM_014653 WSCD2 WSC domain containing 2 -30.48
NM_145290 GPR125 G protein-coupled receptor 125 -29.51
NM_020353 PLSCR4 phospholipid scramblase 4 -28.02
NM_001099921 MAGEB16 melanoma antigen family B, 16 -27.19
NM_033306 CASP4 caspase 4, apoptosis-related cysteine peptidase -23.01
NM_004126 GNG11 guanine nucleotide binding protein (G protein), gamma 11 -22.73
NM_144722 SPEF2 sperm flagellar 2 -20.86
NM_015660 GIMAP2 GTPase, IMAP family member 2 -19.99
NR_027755 LINC00922 long intergenic non-protein coding RNA 922, long non-coding RNA -19.17
NM_018437 HEMGN hemogen -18.55
NM_001005285 OR2AT4 olfactory receptor, family 2, subfamily AT, member 4 -18.19
NM_000537 REN renin -17.26
NM_000519 HBD hemoglobin, delta -16.75
NM_213674 TPM2 tropomyosin 2 (beta) -16.59
NM_002421 MMP1 matrix metallopeptidase 1 -12.23
NM_000361 THBD thrombomodulin -11.98
NM_005807 PRG4 proteoglycan 4 -11.81
NM_080429 AQP10 aquaporin 10 -11.33
NM_139022 TSPAN32 tetraspanin 32 -10.78
NM_024711 GIMAP6 GTPase, IMAP family member 6 -10.55
NM_002145 HOXB2 homeobox B2 -10.22
NM_019032 ADAMTSL4 ADAMTS-like 4 -9.71
NM_002838 PTPRC Protein tyrosine phosphatase, receptor type C -7.81
NR_026812 RUNX1-IT1 RUNX1 intronic transcript 1 -5.91
NM_003153 STAT6 signal transducer and activator of transcription 6 -4.07

Table 1(c).

Most up- and down-regulated genes in TSA+5-Aza treated MV4-11

Gene Bank Accession Gene symbol Gene description ( Homo sapiens) *Folds
change
NM_001145191 FAM200B Family with sequence similarity 200, member B 521.92
NM_197958 LARP6 La ribonucleoprotein domain family, member 6 506.68
NM_017702 DEF8 differentially expressed in FDCP 8 homolog 268.16
NR_027028 GUSBP1 Homo sapiens glucuronidase, beta pseudogene 1 243.94
NM_032905 RBM17 RNA binding motif protein 17 160.05
NM_014773 KIAA0141 KIAA0141 (KIAA0141) 157.47
NM_001204054 NDUFC2 NADH dehydrogenase (ubiquinone) 1, subcomplex unknown 2 155.54
NM_016256 NAGPA N-acetylglucosamine-1-phosphodiester alpha-N-acetylglucosaminidase 141.82
NM_032704 TUBA1C tubulin, alpha 1c 139.42
NM_013268 LGALS13 lectin, galactoside-binding, soluble 13 132.17
NM_004187 KDM5C lysine (K)-specific demethylase 5C 116.85
NM_024097 C1orf50 chromosome 1 open reading frame 50 113.21
NM_006321 ARIH2 ariadne RBR E3 ubiquitin protein ligase 2 97.43
NM_014035 SNX24 sorting nexin 24 94.35
NM_000600 IL6 interleukin 6 (interferon, beta 2) 91.55
NM_138433 KLHDC7B kelch domain containing 7B 89.54
NM_033028 BBS4 Bardet-Biedl syndrome 4 87.94
NM_177424 STX12 syntaxin 12 87.27
NM_015046 SETX senataxin 87.24
NM_001031713 MCUR1 mitochondrial calcium uniporter regulator 1 85.70
NM_001010893 SLC10A5 solute carrier family 10, member 5 79.58
NM_031925 TMEM120A transmembrane protein 120A 78.16
NM_006945 SPRR2D small proline-rich protein 2D 71.36
NM_052936 ATG4A Homo sapiens autophagy related 4A, cysteine peptidase 70.34
NM_014945 ABLIM3 actin binding LIM protein family, member 3 68.78
NM_015701 ERLEC1 endoplasmic reticulum lectin 1 61.29
NM_004672 MAP3K6 mitogen-activated protein kinase kinase kinase 6 59.79
NM_006415 SPTLC1 serine palmitoyltransferase, long chain base subunit 1 59.76
NM_001025300 RAB12 RAB12, member RAS oncogene family 59.16
NM_005988 SPRR2A small proline-rich protein 2A 58.97
NM_001080541 MGA Homo sapiens MGA, MAX dimerization protein 56.75
NM_144569 SPOCD1 Homo sapiens SPOC domain containing 1 54.22
NM_018357 LARP6 Homo sapiens La ribonucleoprotein domain family, member 6 54.17
NM_206818 OSCAR osteoclast associated, immunoglobulin-like receptor 53.30
NM_017956 TRMT12 tRNA methyltransferase 12 homolog (S. cerevisiae) 52.10
NM_005614 RHEB Ras homolog enriched in brain 50.16
NM_012337 CCDC19 coiled-coil domain containing 19 50.03
NM_014884 SUGP2 SURP and G patch domain containing 2 47.37
NM_015335 MED13L mediator complex subunit 13-like 47.11
NM_173698 FAM133A family with sequence similarity 133, member A -153.62
NM_145290 GPR125 G protein-coupled receptor 125 -78.33
NM_017521 FEV Homo sapiens FEV -77.72
NM_001541 HSPB2 Homo sapiens heat shock 27kDa protein 2 -67.21
NM_032501 ACSS1 Homo sapiens acyl-CoA synthetase short-chain family member 1 -63.80
NM_021992 TMSB15A thymosin beta 15a -55.18
NM_012449 STEAP1 six transmembrane epithelial antigen of the prostate 1 -44.93
NM_017414 USP18 ubiquitin specific peptidase 18 -44.70
NM_001803 CD52 CD52 molecule -44.63
NM_004126 GNG11 guanine nucleotide binding protein (G protein), gamma 11 -42.81
NM_000519 HBD hemoglobin, delta -40.08
NM_033258 GNG8 guanine nucleotide binding protein (G protein), gamma 8 -38.65
NM_138444 KCTD12 potassium channel tetramerization domain containing 12 -35.88
NM_002866 RAB3A RAB3A, member RAS oncogene family -35.15
NM_014697 NOS1AP nitric oxide synthase 1 (neuronal) adaptor protein -35.11
NM_018437 HEMGN hemogen -34.39
NM_207459] TEX19 testis expressed 19 -33.52
NM_004982 KCNJ8 potassium inwardly-rectifying channel, subfamily J, member 8 -33.13
NM_013251 TAC3 tachykinin 3 222335545766788WWSSF BBGTT -30.44
NM_032333 FAM213A family with sequence similarity 213, member A -29.38
NM_213599 ANO5 anoctamin 5 -29.37
NM_130776 XAGE3 X antigen family, member 3 -28.64
NM_002585 PBX1 pre-B-cell leukemia homeobox 1 -28.42
NM_001110199 SRRM3 Homo sapiens serine/arginine repetitive matrix 3 -28.20
NM_000537 REN renin -27.47

*Folds-change of treatment group compared to control analyzed by Genespring software analysis, Moderated T-test, p<0.05)

Table 2(a).

Most up- and down-regulated genes in TSA treated Kasumi 1

Gene Bank Accession Gene symbol Gene description
( Homo sapiens)
*Folds
change
NM_139314 ANGPTL4 angiopoietin-like 4 791.26
NM_182908 DHRS2 dehydrogenase/reductase (SDR family) member 2 612.16
NM_001069 TUBB2A tubulin, beta 2A class IIa 574.87
NM_001080434 LMTK3 lemur tyrosine kinase 3 356.19
NM_138345 VWA5B2 von Willebrand factor A domain containing 5B2 331.00
NM_030630 HID1 HID1 domain containing 331.00
NM_006928 PMEL premelanosome protein 323.68
NM_145056 DACT3 dishevelled-binding antagonist of beta-catenin 3 269.03
NM_144698 ANKRD35 ankyrin repeat domain 35, 258.42
NM_014971 EFR3B EFR3 homolog B (S. cerevisiae) 248.79
NM_004933 CDH15 cadherin 15, type 1, M-cadherin (myotubule) 221.35
NM_006086 TUBB3 tubulin, beta 3 class III 205.73
NM_000088 COL1A1 collagen, type I, alpha 1 122.33
NM_017577 GRAMD1C GRAM domain containing 1C 109.67
NM_080860 RSPH1 radial spoke head 1 homolog 109.55
NM_003835 RGS9 regulator of G-protein signaling 9 103.85
NM_001098722 GNG4 guanine nucleotide binding protein (G protein), gamma 4 102.41
NM_005325 HIST1H1A histone cluster 1, H1a 99.67
NM_018667 SMPD3 sphingomyelin phosphodiesterase 3, neutral membrane (neutral sphingomyelinase II) 98.71
NM_033103 RHPN2 rhophilin, Rho GTPase binding protein 2 91.75
NM_007224 NXPH4 neurexophilin 4 88.57
NM_014226 MOK MOK protein kinase 73.56
NM_001077621 VPS37D vacuolar protein sorting 37 homolog D 69.03
NM_001145028 PALM3 paralemmin 3 66.97
NM_177403 RAB7B RAB7B, member RAS oncogene family -264.07
NM_005574 LMO2 Homo sapiens LIM domain only 2 (rhombotin-like 1) -215.33
NM_001004196 CD200 CD200 molecule -162.39
NM_001146 ANGPT1 angiopoietin 1 -159.45
NM_003474 ADAM12 ADAM metallopeptidase domain 12 -137.13
NM_003942 RPS6KA4 Homo sapiens ribosomal protein S6 kinase, 90kDa, polypeptide 4 -136.39
NM_080588 PTPN7 protein tyrosine phosphatase, non-receptor type 7 -133.96
NM_130782 RGS18 regulator of G-protein signaling 18 -119.12
NM_033101 LGALS12 lectin, galactoside-binding, soluble, 12 -94.20
NM_002005 FES FES proto-oncogene, tyrosine kinase -93.71
NM_080387 CLEC4D C-type lectin domain family 4, member D -93.00
NM_024888 LPPR3 lipid phosphate phosphatase-related protein type 3 -80.70
NM_012252 TFEC transcription factor EC -77.90
NM_001805 CEBPE CCAAT/enhancer binding protein (C/EBP), epsilon -69.46
NM_014682 ST18 suppression of tumorigenicity 18, zinc finger -67.63
NM_002467 MYC v-myc avian myelocytomatosis viral oncogene homolog -65.46
NM_005263 GFI1 growth factor independent 1 transcription repressor -64.45
NM_153615 RGL4 ral guanine nucleotide dissociation stimulator-like 4 -63.06
NM_002287 LAIR1 leukocyte-associated immunoglobulin-like receptor 1 -59.78
NM_002586 PBX2 pre-B-cell leukemia homeobox 2 -58.11
NM_005211 CSF1R colony stimulating factor 1 receptor -55.40
NM_002831 PTPN6 protein tyrosine phosphatase, non-receptor type 6 -52.38
NM_000442 PECAM1 platelet/endothelial cell adhesion molecule 1 -52.24

*Folds-change of treatment group compared to control analyzed by Genespring software analysis, Moderated T-test, p<0.05)

Table 2(b).

Most up- and down-regulated genes in 5-Aza treated Kasumi 1

Gene Bank Accession Gene symbol Gene description
( Homo sapiens)
*Folds
change
NM_021120 DLG3 discs, large homolog 3 (Drosophila) 14.12
NM_033114 ZCRB1 zinc finger CCHC-type and RNA binding motif 1 12.82
NM_001110514 EBF4 early B-cell factor 4 12.63
NM_013271 PCSK1N proprotein convertase subtilisin/kexin type 1 inhibitor 11.11
NM_003278 CLEC3B C-type lectin domain family 3, member B 9.44
NM_003456 ZNF205 zinc finger protein 205 9.23
NM_005252 FOS FBJ murine osteosarcoma viral oncogene homolog 8.83
NM_002840 PTPRF protein tyrosine phosphatase, receptor type F 8.83
NM_019058 DDIT4 DNA-damage-inducible transcript 4 8.17
NM_002728 PRG2 proteoglycan 2, bone marrow 7.82
NM_001122962 SIRPB2 signal-regulatory protein beta 2 7.78
NM_001039580 MAP9 microtubule-associated protein 9 7.46
NM_080863 ASB16 ankyrin repeat and SOCS box containing 16 7.21
NM_021158 TRIB3 tribbles pseudokinase 3 6.95
NM_153334 SCARF2 scavenger receptor class F member 2 6.80
NM_002390 ADAM11 ADAM metallopeptidase domain 11 5.63
NM_032797 AIFM2 apoptosis-inducing factor, mitochondrion-associated 2 4.98
NM_004626 WNT11 wingless-type MMTV integration site family, member 11 4.90
NM_032271 TRAF7 TNF receptor-associated factor 7, E3 ubiquitin protein ligase 3.67
NM_001015053 HDAC5 histone deacetylase 5 3.67
NM_001069 TUBB2A tubulin, beta 2A class IIa 2.67
NM_139314 ANGPTL4 angiopoietin-like 4 2.67
NM_002831 PTPN6 protein tyrosine phosphatase, non-receptor type 6 2.27
NM_001292030 TTC39C tetratricopeptide repeat domain 39C -70.59
NM_002844 PTPRK protein tyrosine phosphatase, receptor type K -32.81
NM_198481 VSTM1 V-set and transmembrane domain containing 1 -32.49
NM_000099 CST3 cystatin C -26.47
NM_001244008 KIF1A kinesin family member 1A -22.49
NM_001190467 PRR36 proline rich 36 -21.97
NM_024422 DSC2 desmocollin 2 -20.96
NM_001282735 SPATS2L spermatogenesis associated, serine-rich 2-like -18.59
NM_015238 WWC1 WW and C2 domain containing 1 -16.52
NM_021199 SQRDL sulfide quinone reductase-like (yeast) -15.53
NM_001838 CCR7 chemokine (C-C motif) receptor 7 -13.97
NM_000474 TWIST1 twist family bHLH transcription factor 1 -13.27
NM_012395 CDK14 cyclin-dependent kinase 14 -13.19
NM_000168 GLI3 GLI family zinc finger 3 -12.65
NM_024940 DOCK5 dedicator of cytokinesis 5 -11.91
NM_030906 STK33 serine/threonine kinase 33 -11.90
NM_001900 CST5 cystatin D -11.86
NM_006897 HOXC9 homeobox C9 -11.74
NM_005855 RAMP1 receptor (G protein-coupled) activity modifying protein 1 -11.55
NM_033292 CASP1 caspase 1, apoptosis-related cysteine peptidase -11.50
AK027605 CYP2S1 cytochrome P450, family 2, subfamily S, polypeptide 1 -11.02
NM_003474 ADAM12 ADAM metallopeptidase domain 12 -7.681
NM_172217 IL16 interleukin 16 -4.46
NM_001025300 RAB12 RAB12, member RAS oncogene f -4.89

*Folds-change of treatment group compared to control analyzed by Genespring software analysis, Moderated T-test, p<0.05)

Table 2(c).

Most up- and down-regulated genes in TSA+5-Aza treated Kasumi 1

Gene Bank Accession Gene symbol Gene description ( Homo sapiens) *Folds change
NM_182908 DHRS2 dehydrogenase/reductase (SDR family) member 2 758.66
NM_001080434 LMTK3 lemur tyrosine kinase 3 541.34
NM_001069 TUBB2A tubulin, beta 2A class IIa 435.79
NM_139314 ANGPTL4 angiopoietin-like 4 429.60
NM_138345 VWA5B2 von Willebrand factor A domain containing 5B2 398.46
NM_030630 HID1 HID1 domain containing 341.01
NM_006928 PMEL premelanosome protein 282.05
NM_014971 EFR3B EFR3 homolog B (S. cerevisiae) 263.45
NM_144698 ANKRD35 ankyrin repeat domain 35 220.61
NM_145056 DACT3 dishevelled-binding antagonist of beta-catenin 219.77
NM_004933 CDH15 cadherin 15, type 1, M-cadherin 190.60
NM_006086 TUBB3 tubulin, beta 3 class III 173.87
NM_001098722 GNG4 guanine nucleotide binding protein (G protein), gamma 4 167.50
NM_080860 RSPH1 radial spoke head 1 homolog (Chlamydomonas) 146.52
NM_003835 RGS9 regulator of G-protein signaling 9 126.58
NM_007224 NXPH4 neurexophilin 4 124.19
NM_020770 CGN cingulin 118.29
NM_001145028 PALM3 paralemmin 3 114.39
NM_000088 COL1A1 collagen, type I, alpha 1 111.63
NM_003933 BAIAP3 BAI1-associated protein 3 107.26
NM_017577 GRAMD1C GRAM domain containing 1C 95.72
NM_052899 GPRIN1 G protein regulated inducer of neurite outgrowth 1 95.72
NM_005325 HIST1H1A histone cluster 1, H1a 95.08
NM_033141 MAP3K9 mitogen-activated protein kinase kinase kinase 9 92.48
NM_198573 ENHO energy homeostasis associated 92.06
NM_001039570 KREMEN1 kringle containing transmembrane protein 1 91.54
NM_018667 SMPD3 sphingomyelin phosphodiesterase 3 91.24
NM_012253 TKTL1 transketolase-like 1 87.98
NM_002599 PDE2A phosphodiesterase 2A, cGMP-stimulated 84.11
NM_033259 CAMK2N2 calcium/calmodulin-dependent protein kinase II inhibitor 2 80.49
NM_014226 MOK MOK protein kinase 79.66
NM_001678 ATP1B2 ATPase, Na+/K+ transporting, beta 2 polypeptide 78.33
NM_006500 MCAM melanoma cell adhesion molecule 75.94
NM_001077621 VPS37D vacuolar protein sorting 37 homolog D 74.87
NM_052924 RHPN1 rhophilin, Rho GTPase binding protein 1 74.59
NM_020127 TUFT1 tuftelin 1 73.36
NM_001040709 SYPL2 synaptophysin-like 2 70.97
NM_032432 ABLIM2 actin binding LIM protein family, member 2 70.76
NM_001024401 SBK1 SH3 domain binding kinase 1 68.42
NM_022742 CCDC136 coiled-coil domain containing 136 68.41
NM_021979 HSPA2 heat shock 70kDa protein 2 67.51
NM_000142 FGFR3 fibroblast growth factor receptor 3 65.65
NM_033103 RHPN2 rhophilin, Rho GTPase binding protein 2 65.01
NM_198196 CD96 CD96 molecule (CD96) -228.86
NM_001972 ELANE elastase, neutrophil expressed -172.59
NM_001244008 KIF1A kinesin family member 1A -171.82
NM_133374 ZNF618 zinc finger protein 618 -169.32
NM_020125 SLAMF8 SLAM family member 8 -158.07
NM_003974 DOK2 docking protein 2 -153.14
NM_080387 CLEC4D C-type lectin domain family 4, member D -143.62
NM_130782 RGS18 regulator of G-protein signaling 18 -110.02
NM_033101 LGALS12 lectin, galactoside-binding, soluble, 12 -107.48
NM_178443 FERMT3 fermitin family member 3 -106.90
NM_012072 CD93 CD93 molecule -102.56
NM_001946 DUSP6 dual specificity phosphatase 6 -98.76
NM_012252 TFEC transcription factor EC -92.29
NM_002467 MYC v-myc avian myelocytomatosis viral oncogene homolog -91.05
NM_001004196 CD200 CD200 molecule -87.76
NM_005814 GPA33 glycoprotein A33 (transmembrane) -82.88
NM_153615 RGL4 ral guanine nucleotide dissociation stimulator-like 4 -81.77
NM_080588] PTPN7 protein tyrosine phosphatase, non-receptor type 7 -79.77
NM_014795 ZEB2 zinc finger E-box binding homeobox 2 -79.47
NM_005211 CSF1R colony stimulating factor 1 receptor -74.06
NM_001146 ANGPT1 angiopoietin 1 -70.80
NM_006418 OLFM4 olfactomedin 4 -70.64
NM_014682 ST18 Homo sapiens suppression of tumorigenicity 18 -68.89
NM_177403 RAB7B RAB7B, member RAS oncogene family -67.90
NM_198481 VSTM1 V-set and transmembrane domain containing 1 -66.89
NM_005187 CBFA2T3 core-binding factor, runt domain, alpha subunit 2; translocated to, 3 -61.51
NM_003474 ADAM12 ADAM metallopeptidase domain 12 -59.66
NM_005574 LMO2 LIM domain only 2 -58.27
NM_080387 CLEC4D C-type lectin domain family 4, member D -54.65
NM_001805 CEBPE CCAAT/enhancer binding protein (C/EBP), epsilon -48.73

*Folds-change of treatment group compared to control analyzed by Genespring software analysis, Moderated T-test, p<0.05)

Identification of an optimal Gene Ontology (GO) and KEGG pathway by DAVID software

GO analysis identified 13 optimal GO terms in MV4-11 after TSA, 5-Aza and TSA+5-Aza treatments constituted of 7 highly enriched biological processes (BP); Actin filament organization, Cytoskeleton organization, JAK-STAT, Blood coagulation, Positive regulation of activated T cell proliferation, Positive regulation of MAPK cascade and Cytoskeleton-dependent intracellular transport, related to 6 enriched molecular function (MF); GTPase activity, GTP binding, Structural constituent of cytoskeleton, Signal transducer activity, Polysaccharide binding, and Insulin-like growth factor receptor binding. The transduced GO terms were correspondent to 4 enriched KEGG pathway, which was Viral carcinogenesis, Hepatitis B, JAK-STAT and Phagosome (Table 3a).

Table 3(a).

Gene ontology (GO) profile after TSA, 5-Aza and TSA+5-Aza treatments in MV4-11

GO IDs
GO term
Genes
p-value
Biological processes
GO:0007015 Actin filamen organization ARHGAP6, SPTA1, TPM2, TMSB15A 0.0084
GO:0007010 Cytoskeleton organization ABLIM3, TUBA1C, ANK1, TSPAN32, TUBB3 0.014
GO:0007259 JAK-STAT cascade NMI, STAT5A, SOCS3 0.015
GO:0007596 Blood coagulation CYP4F2, HBD, NFE2, THBD, TFPI 0.022
GO:0042102 Positive regulation of activated T cell proliferation CD24, IGF2, IL6 0.047
GO:0043410 positive regulation of MAPK cascade TIMP2, IGF2, IL6 0.080
GO:0030705 Cytoskeleton-dependent intracellular transport KIF5C, TUBA1C 0.099
Molecular Functions
GO:0003924 GTPase activity GNG11, GNG8, RHEB, RAB3A, TUBA1C, TUBB3 0.010
GO:0005525 GTP binding GIMAP2, GIMAP6, RAB12, RAB3A, RHEB, TUBA1C, TUBB3 0.021
GO:0005200 Structural constituent of cytoskeleton ANK1, SPTA1, TUBA1C, TUBB3 0.024
GO:0004871 Signal transducer activity CD24, GNG11, GNG8, STAT5A, STAT6 0.028
GO:0030247 Polysaccharide binding ENPP3, PRG4 0.076
GO:0005159 Insulin-like growth factor receptor binding IGF2, REN
0.081
Pathways
Viral carcinogenesis CCNA1, HDAC7, HIST1H2BN, STAT5A 0.069
Hepatitis B CCNA1, IL6, STAT5A, STAT6 0.084
JAK-STAT SOCS3, IL6, STAT5A, STAT6 0.084
Phagosome STX12, TUBA1C, TUBB3 0.10

(DAVID software analysis, EASE score 0.1, Benjamini p<0.1)

In Kasumi 1, 16 optimal GO terms by BP were identified; Cell adhesion, Leukocyte migration, Bone mineralization, Regulation of G-protein coupled receptor protein signaling pathway, Positive regulation of cell motility, phagocytosis, Peptidyl-tyrosine dephosphorylation, Protein localization to cell surface, Negative regulation of apoptotic process, Protein phosphorylation, Negative regulation of cell death, Hematopoiesis, Negative regulation of cell proliferation, Response to drug, Angiogenesis and Microtubule-based process, related to 8 MF; Protein tyrosine phosphatase activity, Transmembrane receptor protein tyrosine phosphatase activity, Carbohydrate-binding, Protein kinase activity, Heparin-binding, Protein serine/threonine kinase activity, Beta-catenin binding and Transcription factor binding. The most optimal KEGG pathway induced in Kasumi 1 were; Transcriptional misregulation in cancer, MAPK signaling pathway, PI3K-Akt signaling pathway, Pathways in cancer, Hippo signaling pathway, Proteoglycans in cancer, Ras signaling and Phagosome (Table 3b).

Table 3(b).

Gene ontology (GO) profile after TSA, 5-Aza and TSA+5-Aza treatments in Kasumi 1

GO IDs
GO term
Genes
P-value
Biological processes
GO:0007155 Cell adhesion ADAM12, CDH15, COL1A1, PTPRK, PTPRF, DSC2, ATP1B2, CD96, DSC2, COL1A1, MCAM 0.00093
GO:0050900 Leukocyte migration ANGPTL1, COL1A1, ATP1B2, PECAM1, PTPN6, DOK2 0.0013
GO:0030282 Bone mineralization CLEC3B, WNT11, FGFR3, TUFT1 0.0014
GO:0008277 Regulation of G-protein coupled receptor protein signaling pathway GNG4, RGS18, RGS9,
RAMP1
0.0022
GO:2000147 Positive regulation of cell motility CCR7, CSF1R, TWIST1 0.0037
GO:0006909 Phagocytosis CEBPE, CD93, ELANE, PECAM1 0.0039
GO:0035335 Peptidyl-tyrosine dephosphorylation PTPN6, PTPN7, PTPRK,PTPRF, DUSP6 0.0042
GO:0034394 Protein localization to cell surface WNT11, ANGPTL1, PTPRK 0.0051
GO:0043066 Negative regulation of apoptotic process GLI3, WNT11, ANGPTL1, ANGPTL4, CSF1R, DHRS2, TWIST1, MYC 0.0068
GO:0006468 Protein phosphorylation FES, MOK, WNT11, CDK14, LMTK3, TRIB3, RPS6KA4 0.024
GO:0060548 Negative regulation of cell death WNT11, CST3, MYC 0.030
GO:0030097 Hematopoiesis ANGPTL1, CSF1R, GFI1 0.034
GO:0008285 Negative regulation of cell proliferation PTPN6, PTPRK, GL13, CSF1R, DHRS2, DLG3CBFA2T3 0.048
GO:0042493 Response to drug FOS, COL1A1, CST3, HDAC5, MYC 0.062
GO:0001525 Angiogenesis ANGPTL1, ANGPTL4, PECAM1, RAMP1, MCAM 0.096
GO:0007017 Microtubule-based process TUBB2A, TUBB3 0.10
Molecular Functions
GO:0004725 Protein tyrosine phosphatase activity PTPN6, PTPN7, PTPRF, PTPRK, DUSP6 0.0038
GO:0005001 Transmembrane receptor protein tyrosine phosphatase activity PTPN6, PTPRF, PTPRK 0.0051
GO:0030246 Carbohydrate binding CLEC3B, CLEC4B, PRG2, LGALS12 0.036
GO:0004672 Protein kinase activity MOK, TRIB3, CDK14, LMTK3, STK33, MAP3K9 0.078
GO:0008201 Heparin binding CLEC3B, ELANE, PTPRF, PRG2 0.081
GO:0004674 protein serine/threonine kinase activity MOK, SBK1, LMTK3, MAP3K9, RPS6KA4, STK33 0.091
GO:0008013 Beta-catenin binding GLI3, DACT3, PTPRK 0.095
GO:0003700 Transcription factor binding FOS, PBX2, HDAC5, TWIST1, MYC 0.100
Pathways
Transcriptional misregulation in cancer CEBPE, LMO2, CSF1R, CDK14, MYC, ELANE 0.0014
MAPK signaling pathway FOS, PTPN7, MYC, RPS6KA4 0.010
PI3K-Akt signaling pathway DDIT4, GNG4, ANGPTL1, COL1A1, CSF1R, FGFR3, MYC 0.041
Pathway in cancer FOS, GNG4, GLI3, WNT11, CSF1R, FGFR3, MYC 0.069
Hippo signaling pathway WWCI, WNT11, MYC, DLG3 0.10
Proteoglycans in cancer WNT11, PTPN6, TWIST1, MYC 0.18
Ras signaling GNG4, ANGPTL4, CSF1R, FGFR3 0.23
Phagosome TUBB2A, TUBB3 0.10

(DAVID software analysis, EASE score, p< 0.1)

Identification of Differentially Expressed Genes by Venn Diagram Configuration

In MV4-11, out of 9 common differentially expressed genes between TSA, 5-Aza and TSA+5-Aza treatments, 8 genes (DEF8, GUSBP1, TUBA1C, NDUFC2, ARIH2, STX12, MAP3K6, and RAB12) were commonly up-regulated, while HEMGN was commonly down-regulated in all treatments. Between TSA and 5-Aza treatments, SOCS3 and HIST1H2BN were commonly up-regulated, but PTPRC, GIMAP2, TPM2, CASP4, RUNX1-IT1, and STAT6 were commonly down-regulated. 16 genes were commonly up-regulated in both 5-Aza and TSA+5-Aza treatments (FAM200B, RBM17, C1orf50, TMEM120A, SETX, NAGPA, MCUR1, BBS4, ATG4A, SUGP2, and RHEB). 5 down-regulated genes in 5-Aza (FAM133A, GPR125, GNG11, REN, and HBD) shared common down-regulation with TSA+5-Aza treatments. No gene in common was differentially expressed between TSA and TSA+5-Aza treatments. 25, 16 and 38 genes were exclusively expressed in TSA, 5-Aza and TSA+5-Aza, respectively as shown in Figure 3(a) (p<0.05).

Figure 3(a).

Figure 3(a)

Venn diagram illustrating the genes commonly and exclusively expressed after TSA, 5-Aza and TSA+5-Aza treatments in MV4-11 (adhered to gene selection criteria).

In Kasumi 1, there were 3 common differentially expressed genes across all treatments; 2 genes (ANGPTL4 and TUBB2A) and 1 gene (ADAM12) were commonly up-regulated and down-regulated, respectively. Whereas PTPN6 was either up-regulated in 5-Aza treatment or down-regulated in TSA. VSTM1 and KIF1A were commonly down-regulated in 5-Aza and TSA+5-Aza treatments. There were 36 genes commonly expressed in TSA and TSA+5-Aza treatments with 20 up-regulated and 16 down-regulated genes. 7, 41 and 31 genes were exclusively expressed in TSA, 5-Aza and TSA+5-Aza, respectively as shown in Figure 3(b) (p<0.05).

Figure 3(b).

Figure 3(b)

Venn diagram illustrating the genes commonly and exclusively expressed after TSA, 5-Aza and TSA+5-Aza treatments in Kasumi 1(adhered to gene selection criteria).

Quantitative real-time PCR (qRT-PCR)

To verify the expression of genes, commonly up-regulated genes; SOCS3, TUBA1C, CCNA1, and MAP3K6 in MV4-11; ANGPTL4 and TUBB2A in Kasumi-1, and commonly down-regulated genes; STAT6, PTPRC and RUNX1 in MV4-11, ADAM12 and differentially expressed gene, PTPN6 in Kasumi 1 were selected for validation by qRT-PCR. The results were consistent with that of microarray in both MV4-11 and Kasumi 1 cell lines except for MAP3K6 in MV4-11 (Figure 4).

Figure 4.

Validation of expression levels of selected genes by qRT- PCR

Figure 4

The qRT-PCR results revealed a significant up- and down regulation of several genes in MV4-11 and Kasumi 1 treated with TSA and 5-Aza compared to non-treated cell lines. GAPDH and B2M were used as endogenous controls to which the expression was normalized. Shown in the bar graph is the standard error (SE) of duplicated samples.

Discussion

It was recognized that epigenetic changes serve as a mediator in cancer progression by the changes of gene expression. Epigenetic alterations are reported to concurrently disrupt the essential signaling pathway predisposed cell to uncontrolled growth, longer survival, and metastasis   14 . Histone modifications and DNA hypermethylation are two known epigenetic mechanisms that largely impact the regulation of gene transcription. Histone modification by acetylation has been found to be significantly deficient in acute leukemia patients, compared with the normal individual   15 . In this study, TSA acts by increasing the acetylation level by inhibiting HDAC activity in human leukemic cell lines. Histone acetylation is known to enhance the expression of specific genes that elicit extensive cellular morphology and metabolic changes, such as growth arrest, differentiation, and apoptosis   16 .

Aberrant DNA methylation was the most common epigenetic alteration in leukemia in which an increased level of DNA methylation was observed in AML at remission        17 . 5-Aza reverts DNA methylation to induce antineoplastic activity either by global hypomethylation and direct cytotoxicity on abnormal hematopoietic cells in the bone marrow   18 . 5-Aza inhibits DNMT thus to induce re-expression of the silenced genes to halt tumor growth, and to cause modest differentiation in transformed leukemic cell lines and primary AML   19 . The current study found that both TSA and 5-Aza inhibit the growth of MV4-11 and Kasumi 1 cell lines in a dose-dependent manner. The IC50 of both treatments at 24 hours were lower in MV4-11, compared to Kasumi 1 which could suggest the inhibitory effect of the drugs were less sensitive in Kasumi 1 harboring t(8;21) than in MV4-11 with FLT3-ITD mutation. The variation in the IC50 values would also represent different expression signature in response to TSA and 5-Aza treatments.

It is proposed that the genes which were commonly expressed within TSA, 5-Aza and TSA+5-Aza treatments were epigenetically regulated and involved in the pathogenesis of AML and may serve as candidates for potential biomarkers although they did not share similar GO profile and targeted different signaling pathways. DEF8, NDUFC2, GUSBP1, ARIH2, STX12 and HIST1H2BN were highly re-expressed (more than 100 folds) in either treatment of MV4-11, have not been previously discussed on their role in cancer except for HIST1H2BN. DEP8 is located at chromosome 16 encodes for an activator of intracellular signal transduction reported to carry single nucleotide polymorphism (SNP) rs4268748 at 16q24 with significantly associated with cell cycle regulator, CDK10 expression   20 . GUSBP1 which was located at chromosome 5 were involved in transcriptional regulation by putative alternative promoters (PAPs)        21 . ARIH2 primarily functions in neuronal differentiation was found to be tumor-specific in Glioblastoma multiforme (GBM) correlated with growth suppression in GBM cell lines   22 . Treatment with 5-aza-2′-deoxycytidine resulted in gene re-expression of HIST1H2BN in malignant ovarian cancer   23 . Differential down-regulation of HIST1H2BN was observed in meningiomas was associated with malignant progression   24 . RAB12 is a member of RAS oncogene family, function as small GTPase for intracellular protein transport, activated in stimulus-dependent pattern and promote microtubules-dependent of the cell secretary-granule in mast cell   25  and its up-regulation has been linked with colorectal cancer   26 .

The most optimal GO in MV4-11 were Cytoskeleton organization involving TUBA1C, JAK-STAT cascade involving SOCS3 and STAT6 and the cell cycle involving CCNA1, associated with Phagosome, JAK-STAT pathway and Viral carcinogenesis, respectively, CCNA1 was expressed after TSA treatment with high fold-change (298.44) in MV4-11, but was slightly re-expressed at a low level in 5-Aza and combination treatment (fold-change: 5.67 and 2.81, respectively) (results not shown). CCNA1, located at chromosome 13, encodes for activating regulatory subunit which binds to cyclin-dependent kinases 2 (CDK2) and cell division cycle 2 (CDC2) for the cell cycle machinery to progress into S phase   27 . In normal cells, CCNA1 was prominently expressed in testes, hematopoietic cells, and brain   28 . CCNA1 acts as tumor suppressor gene (TSG) which is epigenetically silenced by hypermethylation in cervical cancer   29 , ovarian, renal and lung carcinoma        30 . In AML, CCNA1 was found to be overexpressed especially in M3 and M2 AML with significant worse overall survival   31 . In addition, upregulation of CCNA1 was observed in leukemic cells in response to DNA damaging agents by increasing DNA repair process   32 . SOCS3, located at chromosome 17 is the known mediators in the JAK-STAT pathway which is strongly related to AML pathogenesis due to its function in blood lineage differentiation, apoptosis, and proliferation        33 . SOCS1, SOCS2 and SOCS3 negatively regulate JAK-STAT signaling in AML patients carrying a FLT3-ITD mutation        34 . SOCS3 has been extensively studied for over 20 years for their role in various diseases, especially in cancer. The most widely reported in SOCS3 was aberrant methylation affecting gene expression and protein function. Hypermethylation of promoter region of SOCS3 resulted in gene silencing implicated in cancer pathogenesis including hematological malignancies   35 , prostate cancer   36 , pancreatic cancer   37 , endometrial carcinoma   38 , hepatocellular carcinoma   39  and breast cancer        40 . Other candidate genes convoluted in the JAK-STAT pathway associated with hematological malignancies are STAT6 and RUNX1. TUBA1C, located at chromosome 12 is a member of tubulin family of microtubules ubiquitously expressed in the esophagus, bone marrow, appendix, brain, colon, bladder and placenta   41 . TUBA1C expression was significantly increased in hepatocellular carcinoma (HCC) on both mRNA and protein level, which predict a poor prognosis   42 , reduced expression in breast cancer associated invasive stage   43  and their expression was susceptible to colorectal cancer risk    44 . Cytochrome P450 (CYP4F2) was the highest re-expressed gene in TSA treatment with more than 1000 fold-change in MV4-11. CYP4F2 is a drug-metabolizing enzyme gene reported to have an epigenetic regulatory role with clinical implication   45 . Inhibition of DNMT and histone deacetylase (HDAC) by 5-Aza and TSA induced the demethylation of CYP1A1 and CYP1A2 leading to their up-regulation   46 .

In Kasumi 1, three common differentially expressed genes in either treatments were ANGPTL4, TUBB2A, and ADAM12 associated with angiogenesis, microtubule-based process, and cell-adhesion, respectively. ANGPTL4, located at chromosome 19 encodes a glycosylated, secreted protein containing a fibrinogen-like C-terminal domain, mainly induced by a nuclear receptor protein, peroxisome-proliferator-activated receptor (PPAR)   47 . It is the most studied among ANGPLT family, functions primarily in the regulation of lipid metabolism, glucose homeostasis, and insulin sensitivity   48 . ANGPTL4 has not been previously discussed in the context of AML. However previous studies have reported ANGPTL4 in various cancer types, including breast cancer, colorectal cancer, prostate cancer, hepatocarcinoma, and renal cell carcinoma, suggesting its important roles in cancer cell growth and progression   49 . In the current study, ANGPTL4 was mutually up-regulated in TSA treatment in both MV4-11 and Kasumi 1 cell lines, thus has wide potential for gene-specific therapy in AML. TUBB2A, located at chromosome 6 is another putative gene in AML with cell-specific expression. It forms a class ll beta-tubulin from six families of tubulins, including, alpha, gamma, delta, epsilon and zeta, and their protein may localize in extracellular exosome, cytoplasm and nucleus, involved in small GTPase activity, GTP binding, nucleotide binding acetylation and methylation   50 . Alpha and beta tubulin sub-families were studied for mutational analysis in human brain tumor and malformations was found in TUBB2A affecting the spectrum of "tubulinopathy" phenotypes51, 52. Mutations in TUBB2A were also explored in epilepsy   51 , gastric carcinoma and lung cancer   53  but not hematological malignancies. ADAM12, located at chromosome 10 was over-expression in non-Hodgkin’s lymphoma that lead to accelerate of proliferation and cell-adhesion   54  and was commonly methylated in chronic lymphocytic leukemia        55 . The roles of ADAM12 in leukemia pathogenesis is still obscure and need further study since the expression of this gene was similarly down-regulated in both treatments. PTPN6 (or SHP1) located at chromosome 12 was differentially regulated in TSA and 5-Aza treatments (re-expressed only in 5-Aza but not TSA). Our previous study showed a positive correlation of PTPN6 re-activation due to hypomethylation in MV4-11 that carry a FLT3-ITD mutation after the 5-Aza treatment   56 . PTPN6 expression has been studied in lymphoma, leukemia and other cancers such as breast cancer, ovarian cancer, prostate cancer, and pancreatic cancer        57 , and in hepatocellular carcinoma   58 . PTPN6 is a downstream mediator in the JAK-STAT pathway, and together with SOCS3 they potentially serve as molecular indicators for pathway-targeted therapy in AML. Another example of the methylation-related gene is PRG2. In the Venn diagram, PRG2 was exclusively expressed in 5-Aza treatment, but not in TSA treatment. The differentially expressed PRG2 was reported in three human leukemic cell lines (K562, THP1, and HL-60)   59 . We also previously reported that the expression of PRG2 was restored after 5-Aza treatment in PKC-412 (Midostaurin) resistant leukemic cell line   56 . DHRS2 and LMTK3 were another highly up-regulated genes in TSA treatment in Kasumi 1 with up to 500 fold change. Their up-regulation was due to histone acetylation.

Finally, despite thousands of genes generated by microarray expression profiling, the highly re-expressed and down-expressed genes perceived in this study were thought to be convoluted with epigenetic regulation of gene transcription in AML. Although only several genes were selected for validation by qRT-PCR, there were many other genes as discussed earlier that may have important roles in cancer pathogenesis.

CONCLUSION

In conclusion, we have identified common differently expressed genes that are importants in epigenetic regulation of AML. Our finding also revealed that Phagosome pathway was the most optimal and common in both MV4-11 and Kasumi 1 AML cell lines. Although MV4-11 and Kasumi 1 transduced different optimal signaling pathways in response to drug treatment, it was shown that MV4-11 mainly targeted the genes in the JAK-STAT signaling, while Kasumi 1 targeted the genes in transcriptional misregulation in cancer, PI3K-Akt and MAPK signaling, which are all critical pathways in oncogenesis. These were due to their different molecular characteristics (FLT3-ITD vs t(8;21) AML1-ETO). The data presented here may serve as a preliminary finding and are useful for further study to explore epigenetic involvement in the pathogenesis of AML.

ACKNOWLEDGEMENTS

This study was financially assisted by Research University grant (1001/PPSP/813050) and Bridging grant (304/PPSP/6316146) from Universiti Sains Malaysia.

CONFLICT OF INTEREST

The authors have no conflict of interest.

References

  • 1.Babon J, Nicola NA. The biology and mechanism of action of suppressor of cytokine signaling 3 (SOCS3) Growth Factors. 2012;30(4):207–19. doi: 10.3109/08977194.2012.687375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Arber DA, Orazi A, Hasserji RP, et al. Introduction and overview of the classification of myeloid neoplasms. WHO classification of tumors of haematopoietic and lymphoid tissues. Revised 4th Edition ed. Geneva: World Health Organization (WHO) Press; 2017. pp. 172–75. [Google Scholar]
  • 3.American Cancer Society:Cancer Facts &amp; Figures. Atlanta: American Cancer Society; c1913-2019 [updated 20 November 2018] [Accessed 24 October 2018]. American Cancer Society. Available from: https://www.cancer.org/research/cancer-facts-statistics/all-cancer-facts-figures/cancer-facts-figures-2018.html.
  • 4.Pollack JR. A perspective on DNA microarrays in pathology research and practice. Am J Pathol. 2007;171(2):375–85. doi: 10.2353/ajpath.2007.070342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Golub TR, Slonim DK, Tamayo P, et al. Molecular Classification of Cancer: Class Discovery and Class Prediction by Gene Expression Monitoring. Science. 1999;286(5439):531–37. doi: 10.1126/science.286.5439.531. [DOI] [PubMed] [Google Scholar]
  • 6.Kumar CC. Genetic abnormalities and challenges in the treatment of acute myeloid leukemia. Genes Cancer. 2011;2(2):95–107. doi: 10.1177/1947601911408076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Li S, Mason CE, Melnick A. Genetic and epigenetic heterogeneity in acute myeloid leukemia. Curr Opin Genet Dev. 2016;36:100–06. doi: 10.1016/j.gde.2016.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.You JS, Jones PA. Cancer genetics and epigenetics: two sides of the same coin? Cancer Cell. 2012;22(1):9–20. doi: 10.1016/j.ccr.2012.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Sharma S, Kelly TK, Jones PA. Epigenetics in cancer. Carcinogenesis. 2010;31(1):27–36. doi: 10.1093/carcin/bgp220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Baylin SB, Jones PA. A decade of exploring the cancer epigenome - biological and translational implications. Nat Rev Cancer. 2011;11(10):726–34. doi: 10.1038/nrc3130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Hatziapostolou M, Iliopoulos D. Epigenetic aberrations during oncogenesis. Cell Mol Life Sci. 2011;68(10):1681–702. doi: 10.1007/s00018-010-0624-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Nicolas D, Zoller B, Suter DM, et al. Modulation of transcriptional burst frequency by histone acetylation. Proc Natl Acad Sci USA. 2018;115(27):7153–58. doi: 10.1073/pnas.1722330115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Hosack DA, Dennis G Jr, Sherman BT, et al. Identifying biological themes within lists of genes with EASE. Genome Biol. 2003;4(10):R70. doi: 10.1186/gb-2003-4-10-r70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kagohara LT, Stein-O'Brien GL, Kelley D, et al. Epigenetic regulation of gene expression in cancer: techniques, resources and analysis. Brief Funct Genomics. 2018 ;17(1):49–63. doi: 10.1093/bfgp/elx018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Xiao L, Huang Y, Zhen R, et al. Deficient Histone Acetylation in Acute Leukemia and the Correction by an Isothiocyanate. Acta Haematol. 2010;123(2):71–76. doi: 10.1159/000264628. [DOI] [PubMed] [Google Scholar]
  • 16.Shankar S, Srivastava RK. Histone deacetylase inhibitors: mechanisms and clinical significance in cancer: HDAC inhibitor-induced apoptosis. Adv Exp Med Biol. 2008;615:261–98. doi: 10.1007/978-1-4020-6554-5_13. [DOI] [PubMed] [Google Scholar]
  • 17.Agrawal S, Unterberg M, Koschmieder S, et al. DNA methylation of tumor suppressor genes in clinical remission predicts the relapse risk in acute myeloid leukemia. Cancer Res. 2007;67(3):1370–7. doi: 10.1158/0008-5472.CAN-06-1681. [DOI] [PubMed] [Google Scholar]
  • 18.NCI Drug Dictionary: Azacitidine. Bethesda: US National Cancer Institute; [updated 1 August 2018]; [Accessed 25 October 2018]. Available from: https://www.cancer.gov/publications/dictionaries/cancer-drug/def/azacitidine. [Google Scholar]
  • 19.Leone G, D'Alo F, Zardo G, et al. Epigenetic treatment of myelodysplastic syndromes and acute myeloid leukemias. Curr Med Chem. 2008;15(13):1274–87. doi: 10.2174/092986708784534947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Asgari MM, Wang W, Ioannidis NM, et al. Identification of Susceptibility Loci for Cutaneous Squamous Cell Carcinoma. J Invest Dermatol. 2016;136(5):930–37. doi: 10.1016/j.jid.2016.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kimura K, Wakamatsu A, Suzuki Y, et al. Diversification of transcriptional modulation: large-scale identification and characterization of putative alternative promoters of human genes. Genome Res. 2006;16(1):55–65. doi: 10.1101/gr.4039406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Harisankar A. Identification of novel genes with important functions in glioblastoma multiforme and acute myeloid leukemia. Huddinge. Huddinge: Institute för medicine; 2018. [Google Scholar]
  • 23.Liao YP, Chen LY, Huang RL, et al. Hypomethylation signature of tumor-initiating cells predicts poor prognosis of ovarian cancer patients. Hum Mol Genet. 2014;23(7):1894–906. doi: 10.1093/hmg/ddt583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Pérez ME, Rodríguez de LÁ, Ribalta T, et al. Differential expression profiling analyses identifies downregulation of 1p, 6q, and 14q genes and overexpression of 6p histone cluster 1 genes as markers of recurrence in meningiomas. Neuro Oncol. 2010;12(12):1278–90. doi: 10.1093/neuonc/noq081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Efergan A, Azouz NP, Klein O, et al. Rab12 Regulates Retrograde Transport of Mast Cell Secretory Granules by Interacting with the RILP–Dynein Complex. J Immunol. 2016;196(3):1091–101. doi: 10.4049/jimmunol.1500731. [DOI] [PubMed] [Google Scholar]
  • 26.Yoshida T, Kobayashi T, Itoda M, et al. Clinical omics analysis of colorectal cancer incorporating copy number aberrations and gene expression data. Cancer Inform. 2010;9:147–61. doi: 10.4137/cin.s3851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Lapenna S, Giordano A. Cell cycle kinases as therapeutic targets for cancer. Nat Rev Drug Discov. 2009;8(7):547–66. doi: 10.1038/nrd2907. [DOI] [PubMed] [Google Scholar]
  • 28.National Cancer for Biotechnology Information (NCBI) Gene ID: 8900. CCNA1 cyclin A1 [Homo sapiens (human)] Bethesda: U.S. National Library of Medicine; c1988-2019 [updated updated 7 September 2018]; [Accessed 7 October 2018]. Available from: https://www.ncbi.nlm.nih.gov/gene/84790. [Google Scholar]
  • 29.Yang N, Eijsink JJH, Lendvai Á, et al. Methylation Markers for CCNA1 & C13ORF18 Are Strongly Associated with High-Grade Cervical Intraepithelial Neoplasia and Cervical Cancer in Cervical Scrapings. Cancer Epidemiol Biomarkers Prev. 2009;18(11):3000. doi: 10.1158/1055-9965.EPI-09-0405. [DOI] [PubMed] [Google Scholar]
  • 30.Rivera A, Mavila A, Bayless KJ, et al. Cyclin A1 is a p53-induced gene that mediates apoptosis, G2/M arrest, and mitotic catastrophe in renal, ovarian, and lung carcinoma cells. Cell Mol Life Sci. 2006;63(12):1425–39. doi: 10.1007/s00018-006-5521-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ekberg J, Holm C, Jalili S, et al. Expression of cyclin A1 and cell cycle proteins in hematopoietic cells and acute myeloid leukemia and links to patient outcome. Eur J Haematol. 2005;75(2):106–15. doi: 10.1111/j.1600-0609.2005.00473.x. [DOI] [PubMed] [Google Scholar]
  • 32.Federico M, Symonds CE, Bagella L, et al. R-Roscovitine (Seliciclib) prevents DNA damage-induced cyclin A1 upregulation and hinders non-homologous end-joining (NHEJ) DNA repair. Mol Cancer. 2010;9:208. doi: 10.1186/1476-4598-9-208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Vainchenker W, Constantinescu SN. JAK/STAT signaling in hematological malignancies. Oncogene. 2013;32(21):2601–13. doi: 10.1038/onc.2012.347. [DOI] [PubMed] [Google Scholar]
  • 34.Kazi JU, Ronnstrand L. Suppressor of cytokine signaling 2 (SOCS2) associates with FLT3 and negatively regulates downstream signaling. Mol Oncol. 2013;7(3):693–703. doi: 10.1016/j.molonc.2013.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Fourouclas N, Li J, Gilby DC, et al. Methylation of the suppressor of cytokine signaling 3 gene in myeloproliferative disorders. Haematologica. 2008;93(11):1635. doi: 10.3324/haematol.13043. [DOI] [PubMed] [Google Scholar]
  • 36.Pierconti F, Martini M, Pinto F, et al. Epigenetic silencing of SOCS3 identifies a subset of prostate cancer with an aggressive behavior. The Prostate. 2010;71(3):318–25. doi: 10.1002/pros.21245. [DOI] [PubMed] [Google Scholar]
  • 37.Wang J, Zhou H, Han Y. SOCS3 methylation in synergy with Reg3A overexpression promotes cell growth in pancreatic cancer. J Mol Med (Berl) 2014;92(12):1257–69. doi: 10.1007/s00109-014-1184-8. [DOI] [PubMed] [Google Scholar]
  • 38.Chen H, Zhang C, Sheng Y, et al. Frequent SOCS3 and 3OST2 promoter methylation and their epigenetic regulation in endometrial carcinoma. Am J Cancer Res. 2014 Dec 15;5(1):180–90. [PMC free article] [PubMed] [Google Scholar]
  • 39.Zhang X, You Q, Zhang X, et al. SOCS3 Methylation Predicts a Poor Prognosis in HBV Infection-Related Hepatocellular Carcinoma. Int J Mol Sci. 2015;16(9) doi: 10.3390/ijms160922662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Barclay JL, Anderson ST, Waters MJ, et al. SOCS3 as a tumor suppressor in breast cancer cells, and its regulation by PRL. Int J Cancer. 2009;124(8):1756–66. doi: 10.1002/ijc.24172. [DOI] [PubMed] [Google Scholar]
  • 41.National Cancer for Biotechnology Information (NCBI) Gene ID: 84790. TUBA1C tubulin alpha 1c [Homo sapiens (human)] Bethesda: U.S. National Library of Medicine; c1988-2019 [updated 7 September 2018]; [Accessed 7 October 2018]. Available from: https://www.ncbi.nlm.nih.gov/gene/84790. [Google Scholar]
  • 42.Wang J, Chen W, Wei W, et al. Oncogene TUBA1C promotes migration and proliferation in hepatocellular carcinoma and predicts a poor prognosis. Oncotarget. 2017;8(56):96215–24. doi: 10.18632/oncotarget.21894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Chen D, Li Y, Wang L, et al. SEMA6D Expression and Patient Survival in Breast Invasive Carcinoma. Int J Breast Cancer. 2015;2015:539721. doi: 10.1155/2015/539721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Closa A, Cordero D, Sanz-Pamplona R, et al. Identification of candidate susceptibility genes for colorectal cancer through eQTL analysis. Carcinogenesis. 2014;35(9):2039–46. doi: 10.1093/carcin/bgu092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Tang X, Chen S. Epigenetic Regulation of Cytochrome P450 Enzymes and Clinical Implication. Curr Drug Metab. 2015;16(2):86–96. doi: 10.2174/138920021602150713114159. [DOI] [PubMed] [Google Scholar]
  • 46.Park HJ, Choi YJ, Kim JW, et al. Differences in the Epigenetic Regulation of Cytochrome P450 Genes between Human Embryonic Stem Cell-Derived Hepatocytes and Primary Hepatocytes. PLoS One. 2015;10(7):e0132992–e92. doi: 10.1371/journal.pone.0132992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.La Paglia L, Listi A, Caruso S, et al. Potential Role of ANGPTL4 in the Cross Talk between Metabolism and Cancer through PPAR Signaling Pathway. PPAR Res. 2017;2017:8187235. doi: 10.1155/2017/8187235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Genecards Human gene Database (GCID:GC19P008363) ANGPTL4 Gene (Protein Coding) Israel: Weizmann Institute of Science; c1996-2019 [updated 10 September 2018]; [Accessed 4 October 2018]. Available from: https://www.genecards.org/cgi-bin/carddisp.pl?gene=ANGPTL4. [Google Scholar]
  • 49.Tan MJ, Teo Z, Sng MK, et al. Emerging roles of angiopoietin-like 4 in human cancer. Mol Cancer Res. 2012;10(6):677–88. doi: 10.1158/1541-7786.MCR-11-0519. [DOI] [PubMed] [Google Scholar]
  • 50.UniProtKB - Q13885 (TBB2A_HUMAN) Protein knowledgebase (UniProtKB) Bethesda: National Institute of Health; c2002-2019 [updated 16 March 2018] [Accessed 21 August 2018]. Available from: https:// www.uniprot.org/uniprot/Q13885.
  • 51.Cushion Thomas D, Paciorkowski Alex R, Pilz Daniela T, et al. De Novo Mutations in the Beta-Tubulin Gene TUBB2A Cause Simplified Gyral Patterning and Infantile-Onset Epilepsy. Am J Hum Genet. 2014;94(4):634–41. doi: 10.1016/j.ajhg.2014.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Romaniello R, Arrigoni F, Bassi MT, et al. Mutations in α- and β-tubulin encoding genes: Implications in brain malformations. Brain Dev. 2015;37(3):273–80. doi: 10.1016/j.braindev.2014.06.002. [DOI] [PubMed] [Google Scholar]
  • 53.The Human Protein Atlas: TUBB2A. Knut &amp; Alice Wallenberg foundation 2018. [Accessed 24 june 2018]. Available from: https://v18.proteinatlas.org/ENSG00000137267-TUBB2A/tissue.
  • 54.Zhong F, Ouyang Y, Wang Q, et al. Upregulation of ADAM12 contributes to accelerated cell proliferation and cell adhesion-mediated drug resistance (CAM-DR) in Non-Hodgkin’s Lymphoma AU - Yin, Haibing. Hematology. 2017;22(9):527–535. doi: 10.1080/10245332.2017.1312205. [DOI] [PubMed] [Google Scholar]
  • 55.Rahmatpanah FB, Carstens S, Hooshmand SI, et al. Large-scale analysis of DNA methylation in chronic lymphocytic leukemia. Epigenomics. 2009;1(1):39–61. doi: 10.2217/epi.09.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Al-jamal H, Asmaa MJ, Sidek M, et al. Restoration of PRG2 Expression by 5-Azacytidine Involves in Sensitivity of PKC-412 (Midostaurin) Resistant FLT3-ITD Positive Acute Myeloid Leukaemia Cells. J Hematol Thrombo Dis. 2015;3(1):1–7. [Google Scholar]
  • 57.Wu C, Sun M, Liu L, et al. The function of the protein tyrosine phosphatase SHP-1 in cancer. Gene. 2003;306:1–12. doi: 10.1016/s0378-1119(03)00400-1. [DOI] [PubMed] [Google Scholar]
  • 58.Wen LZ, Ding K, Wang ZR, et al. SHP-1 acts as a Tumor Suppressor in Hepatocarcinogenesis and HCC Progression. Cancer Res. 2018;78(16):4680–4691. doi: 10.1158/0008-5472.CAN-17-3896. [DOI] [PubMed] [Google Scholar]
  • 59.Wang H, Hu H, Zhang Q, et al. Dynamic transcriptomes of human myeloid leukemia cells. Genomics. 2013;102(4):250–6. doi: 10.1016/j.ygeno.2013.06.004. [DOI] [PubMed] [Google Scholar]

Articles from International Journal of Hematology-Oncology and Stem Cell Research are provided here courtesy of Tehran University of Medical Sciences

RESOURCES