Inhibin-α gene mutations and mRNA levels in human lymphoid and myeloid leukemia cells

  • Authors:
    • Young Il Kim
    • Seung-Won Park
    • Hye Shin Kwon
    • Hyung-Seok Yang
    • Sun Young Cho
    • Young Jin Kim
    • Hee Joo Lee
  • View Affiliations

  • Published online on: March 2, 2017     https://doi.org/10.3892/ijo.2017.3895
  • Pages: 1403-1412
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The inhibin-α gene was identified as a tumor suppressor gene in the gonads and adrenal glands by functional studies using knockout mice. Methylation of CpG sites within the regulatory regions of tumor suppressor gene is frequently associated with their transcriptional silencing. We investigated epigenetic modifications, changes in loss of heterozygosity (LOH), and mutation of the inhibin-α gene, and regulation of transcriptional expression in response to inhibitors of DNA methylation (5-aza-2'-deoxycytidine, 5-AzaC) in human lymphoid (Jurkat, Molt-4, Raji, and IM-9) and myeloid (HL-60, Kasumi-1, and K562) leukemia cells. The inhibin-α promoter was hypermethylated in lymphoid (Molt-4 and Raji) and myeloid (HL-60 and Kasumi-1) leukemia cells. Inhibin-α gene mutations differed significantly between lymphoid (heterozygote) and myeloid (homozygote) leukemia cells. LOH in the inhibin-α gene was detected in lymphoid and myeloid leukemia cells, with the exception of Jurkat cells. Treatment with 5-AzaC, a demethylating agent, resulted in increased inhibin-α mRNA and protein levels in most of the cell lines. Also, 5-AzaC treatment inhibited cell proliferation and induced apoptosis. Taken together, our results reveal that the inhibin-α gene is transcriptionally silenced in human leukemia cells and that reactivation is suppressed by a demethylating agent. In addition, mutations in, and expression levels of, the inhibin-α gene differed between human lymphoid and myeloid leukemia cells.

Introduction

Methylation of islands has been shown to inhibit transcription directly or stabilize chromatin in a conformation that prevents transcription (1). Hypermethylation of CpG islands is an important epigenetic mechanism for silencing the transcription of many genes (2). DNA methylation inhibits gene transcription by affecting the chromatin structure, in particular via the protein complexes comprising methyl-binding domains, transcriptional corepressors, and histone deacetylase in hypermethylated regions of DNA (35). In hematopoietic development, this was first demonstrated for erythropoiesis, and later also for lymphoid and myeloid differentiation (69). Aberrant methylation of tumor-suppressor and growth-regulatory genes has been reported as the most frequent alteration in both hematologic neoplasms and solid tumors (10). Promoter methylation is an increasingly recognized mechanism of transcriptional silencing in human cancer. Downregulated expression of target genes results from single transitional changes in these important regulatory sequences. It has been demonstrated that promoter methylation can be induced by viral agents or by recruitment of the methyl-transferase enzymatic machinery (11,12).

Inhibins and activins, members of the transforming growth factor-β (TGF-β) superfamily, are polypeptides that were originally isolated from ovarian fluid, based on their effect on pituitary follicle-stimulating hormone (FSH) production and secretion. Inhibins are heterodimers that are composed of a common α subunit and one of two homologous β subunits (βA and βB) (13,14). More recently, both inhibins and activins have been implicated in endocrine-related cancers (15). The inhibin-α gene was identified as a tumor suppressor gene in the gonads and adrenal glands by functional studies using knockout mice (16,17). This has raised the question of whether it plays a broader role as a tumor suppressor outside the reproductive axis. Furthermore, hypermethylation of the inhibin-α gene promoter and loss of heterozygosity (LOH) at 2q32-36, the chromosome region harboring the inhibin-α gene, has been reported in prostate carcinoma and gastric cancer (18,19). Moreover, there was a positive correlation between loss of inhibin expression and malignancy of these human prostate carcinoma cells (20). Hypermethylation of the inhibin-α promoter and LOH are frequently associated with silencing or loss of expression of tumor suppressor genes, and the effects of deletions involving the 2q33-36 regions in human leukemia cells are unclear. Recently many studies have been made regulating promoter methylation which is a useful biomarker for the diagnosis of cancer patients (21,22).

We investigated the epigenetic modifications, changes in LOH and mutations of the inhibin-α gene, and regulation of transcriptional expression by inhibitors of DNA methylation (5-aza-2′-deoxycytidine, 5-AzaC) in human lymphoid and myeloid leukemia cells.

Materials and methods

Cell culture

Lymphoid (Jurkat, Molt-4, IM-9, and Raji) and myeloid (HL-60, Kasumi-1, and K562) human leukemia cell lines were purchased from the American Tissue Culture Collection (ATCC, Rockville, MD, USA). Cells were cultured in RPMI-1640 medium (Gibco, Grand Island, NY, USA) containing 10% fetal bovine serum (FBS), 100 U/ml penicillin and 100 μg/ml streptomycin at 37°C in a humidified atmosphere of 5% CO2 in 95% air.

Bisulfite modification

The methylation status of the promoter CpG islands of the inhibin-α subunit gene was analyzed by methylation specific PCR (MSP) using sodium-bisulfite- converted DNA (23). Genomic DNA was extracted using the Wizard Genomic DNA purification kit (Promega, Madison, WI, USA). DNA (2 μg) in a volume of a 50 μl was denatured with NaOH (final concentration, 0.2 M) and incubated at 37°C for 15 min. Then, 30 μl of 10 mM hydroquinone and 520 μl of 3 M sodium bisulfite (Sigma-Aldrich, St. Louis, MO, USA) at pH 5.0, both freshly prepared, were added and mixed, and samples were incubated under mineral oil at 55°C for 16 h. DNA was then desalted using the Wizard DNA Clean-Up System (Promega), desulfonated by addition of NaOH (final concentration, 0.3 M), and incubated at 37°C for 15 min. The solution was neutralized by addition of ammonium acetate (final concentration, 3.0 M), and the DNA was ethanol- precipitated, dried, re-suspended in 20 μl of water and used immediately or stored at −20°C.

Determination of methylation status

Methylation was assessed by PCR and sequence analysis of bisulfite-treated DNA. The bisulfite reaction converted unmethylated cytosines to uracils, whereas methylated cytosines were unchanged. The inhibin-α subunit 5′-UTR region was amplified by nested PCR using primers for the bisulfite-converted sequence (18). Primers 1 (5′-GATAAGAGTTTAGATTGGTTTTATTGGTT-3′) and 2 (5′-ACACCATAACTCACCTAACCCTACTAATAA-3′) were used for the first round of PCR, and primers 3 (5′-ACCCCTTCTACCAAAATCTACCCAAAA-3′) and 4 (5′-GAAGGTGTTGTATGTTTGTATGTGTGAGTT-3′) were used for the second round. The first round of PCR was performed in 25 μl reactions with 2 μl of bisulfite-converted DNA, 1X PCR buffer [10 mM Tris (pH 8.3), 50 mM KCl, 1.5 mM MgCl2], 200 μM dNTPs, 10 pmol of primers 1 and 2, and 1 U AmpliTaq Gold DNA polymerase (Roche, Applied Biosystems, Foster City, CA, USA). PCR cycles consisted of 95°C for 15 min, followed by 5 cycles of 95°C for 1 min, 50°C for 2 min, and 72°C for 3 min, and then 30 cycles of 95°C for 1 min, 55°C for 2 min, and 72°C for 2 min, with a final incubation step of 72°C for 10 min. A sample of 2 μl from the first PCR was amplified in a 25-μl reaction as above, except that primers 3 and 4 were used. PCR cycling conditions were as for the first reaction, with the exception that the annealing temperature was increased to 60°C. PCR products were gel-purified, ligated into the pCR 2.1 vector, and cloned using the TOPO TA Cloning kit according to the manufacturer's instructions (Invitrogen, Carlsbad, CA, USA). For each PCR, 10 clones were sequenced and methylation at each of the seven CpG sites was determined. Overall percentage methylation of each sample was determined as the mean of the percentage methylation at the seven individual CpG sites.

DNA analysis

DNA was isolated from cultured cells using standard methods. Two regions of the inhibin-α subunit gene were amplified from genomic DNA by PCR using specific oligonucleotide primers (24). The first 240-bp region (fragment A), which includes 140 bp of the 5′-UTR and 100 bp of exon 1, was amplified using the primers AF (5′-GACTGGGGAAGACTGGATGA-3′) and AR (5′-TCACCTTGGCCAGAACAAGT-3′). The second 396-bp region (fragment B), which comprises part of exon 2, was amplified using the primers BF (AGCAGCCTCCAATAGCTCTG-3′) and BR (5′-AGCTCCTGGAAGGAGATGTTC-3′). Genomic DNA (200 ng) was amplified in a 50-μl volume reaction containing 1X PCR buffer, 2 mM MgCl2, 2.5% DMSO, 0.2 mM dNTP, 20 pmol of each specific primer and 1.5 U AmpliTaq Gold DNA polymerase. The amplification conditions were as follows: 35 cycles comprising an initial denaturation at 95°C for 14 min, then denaturation at 95°C for 40 sec, annealing at 57°C for 30 sec, and extension at 72°C for 1 min, followed by a final extension at 72°C for 7 min. Polymorphism −16C>T in 5′-UTR was screened in the samples by restriction enzyme analysis using SpeI (New England Biolabs, Ipswich, MA, USA). Briefly, fragment A was amplified by PCR and 5 μl of purified PCR product was digested overnight at 37°C with 5 U of Spel, electrophoresed in 8% polyacrylamide gels, stained with ethidium bromide and photographed. The presence of the 240-bp fragment indicated a variant homozygous for C, whereas the presence of two fragments of 120 bp corresponded to a variant homozygous for T. Substitution 769G>A of exon 2 was analyzed by digestion of fragment B with appropriate restriction enzymes. Five microliters of purified PCR product was digested overnight at 37°C with 5 U of BsrFI and analyzed as described above. The restriction site, which renders two fragments of 340 and 56 bp, is abolished in the mutated allele. In addition, 5 μl of purified PCR product was digested overnight at 37°C with 5 U of Fnu4HI, electrophoresed in 15% polyacrylamide gels, stained with ethidium bromide and visualized by image analysis. The 396-bp fragment yields four fragments of 153, 107, 51 and 25 bp, among others of lower molecular weight, in the wild-type allele, whereas the allele with substitution 769G>A yields four fragments of 153, 107, 76, and 51 bp, among others of lower molecular weight.

LOH analysis

LOH was determined using microsatellite markers on 2q32-q33 (D2S389) and 2q33-q36 (D2S128), as described previously (18). The primers used were D2S389 (5′-TAAAGCCTAGTGGAAGATCATC-3′, 5′-GCTGAGTTAACAGTTATCAACAATT-3′) and D2S128 (5′-AAACTGAGATTTGTCTAAGGGG-3′, 5′-AGCCAGGAATTTTTGCTATT-3′). PCR was performed in 20 μl reactions consisting of 200 ng of DNA, 1X PCR buffer, 0.2 mM dNTPs, 10 pmol of each primer, and 1 U AmpliTaq Gold DNA polymerase. The amplification conditions were as follows: 35 cycles of an initial denaturation at 95°C for 14 min, a second denaturation at 95°C for 1 min, annealing at 55°C for 1 min, and extension at 72°C for 1 min, followed by a final extension at 72°C for 10 min. Then, 10 μ1 of PCR products was mixed with 10 μ1 of stop solution containing 95% formamide, 10 mM NaOH, 0.25% bromophenol blue, and 0.25% xylene cyanol FF. The mixture was denatured at 95°C for 5 min, placed on ice for 5 min, electrophoresed in 12% polyacrylamide gels containing 10% glycerol with 1X TBE buffer, and stained with ethidium bromide.

5-Aza-2′-deoxycytidine treatment

Cells were seeded at a density of 5×105/100 mm dish, allowed to attach for 24 h and then treated with various concentration of 5-aza-2′-deoxycytidine (5-AzaC, Sigma) for 5 days. The medium and drug were replaced every 2 days. At the end of the treatment period, the medium was removed and the cell pellets were used for analysis.

RNA extraction and real-time PCR

Total RNA was extracted from cultured cells using the TRIzol Reagent kit following the manufacturer's protocol (Invitrogen). First-strand cDNA was synthesized from 1 μg of DNase-treated RNA using a reverse transcription system (Promega) according to the manufacturer's protocol with random hexamers. PCR was performed with 2 μl cDNA in a 25-μl reaction mixture of 1X PCR buffer, 0.2 mM of each dNTP, 10 pmol of primers for inhibin-α (5′-AGGAAGAGGAGGATGTCTCC-3′ and 5′-GAGTAACCTCCATCCCGAGGT-3′; 823 bp), betaglycan (5′-ACATGGATAAGAAGCGATTCAGC-3′ and 5′-AACGCAATGCCCATCACGGTTAG-3′, 331 bp), and β-actin (5′-CTTCTACAATGAGCTGCGTG-3′ and 5′-TCATGAGGTAGTCAGTCAGG-3′; 305 bp), and 1 U AmpliTaq Gold DNA polymerase. The reactions were carried out in a thermal cycler with an initial denaturation step at 95°C for 14 min, followed by 35 cycles (25 cycle for β-actin) of denaturation at 95°C for 1 min, primer annealing at 50°C (inhibin-α) to 55°C (β-actin) for 1 min, and a final extension at 72°C for 1 min. The reaction was terminated at 72°C for 10 min; samples were stored at 4°C. Ten microliters of PCR products were separated by electrophoresis in a 2% agarose gel containing ethidium bromide (0.5 μg/ml) and visualized by image analysis (Gel Doc 1000 Gel Documentation System; Bio-Rad, Hercules, CA, USA). Real-time PCR was performed on a StepOnePlus Real-Time PCR System with Power SYBR Green PCR Master Mix (Applied Biosystems). The gene-specific primer sequences were: inhibin-α, 5′-CTCGGATGGAGGTTACTCTTTCAA-3′ and 5′-GAAGACCCCCCACCCTTAGA-3′ (88 bp); betaglycan, 5′-CAAAGCAGCAGAAGGGTGTGT-3′ and 5′-GGTGATTAGCTCGATGATGTGTACTT-3′ (73 bp); and β-actin, 5′-GCGAGAAGATGACCCAGATC-3′ and 5′-GGATAGCACAGCCTGGATAG-3′ (77 bp). PCR was performed with 1 μl of cDNA in a 20-μl reaction mixture containing 10 μl of Power SYBR Green PCR Master Mix, 2 μl of primers, and 7 μl of PCR-grade water. The reaction conditions were denaturation at 95°C for 10 min, followed by 40 cycles of 95°C for 15 sec and 60°C for 1 min. The crossing points of the target genes with β-actin were calculated using the formula 2−(target gene-β-actin), and relative amounts were quantified.

FITC-flow cytometric analysis of inhibin-α protein

Cultured cells were detached with 0.05% trypsin-EDTA solution. After washing with cold PBS, cells were incubated with a 1:50 dilution of anti-inhibin-α goat polyclonal antibody (Santa Cruz Biotechnology, Santa Cruz, CA, USA) or normal goat serum as a negative control for 30 min at 4°C. After washing three times with cold PBS, cells were stained with a fluorescein isothiocyanate (FITC)-labeled donkey antibody (1:50 dilution) to rabbit immunoglobulin for 30 min at 4°C. Washing was repeated in the same manner and cell-surface immunofluorescence was analyzed using a FACSCalibur instrument together with CellQuest software (Becton-Dickinson, San Jose, CA, USA).

Determination of cell doubling time

Cells were treated with 5 μM 5-AzaC for 5 days and washed with PBS. Cells were seeded at 2×104/ml in 12-well plates containing culture medium, and cell number/dish was determined by trypan blue assay daily for 5 consecutive days. Untreated cells were analyzed under similar conditions as a control. The average cell number from two plates was determined, and the mean cell numbers were plotted to calculate the doubling times. The cell population doubling time was calibrated using the Kuchler formula (25).

Cell cycle analysis

Cells (5×105) were treated with 5 μM 5-AzaC for 5 days. At the end of the treatment period, cells were harvested, washed with PBS, fixed with 70% ethanol for 1 h, treated with RNAsin (20 μg/ml) at 37°C for 1 h, and stained with propidium iodide (50 μg/ml; Sigma). DNA content at each cell cycle stage was analyzed using a FACSCalibur instrument together with CellQuest software (Becton-Dickinson).

Statistical analysis

Values are expressed as means ± SD. Student's t-test was used to evaluate differences among the samples. Statistical analyses were performed using GraphPad Prism 5 software (GraphPad Software Inc., San Diego, CA, USA). *P<0.05 and **P<0.01 were considered to indicate statistical significance.

Results

Methylation status of the inhibin-α gene promoter in human leukemia cells

Methylation at the seven CpG sites, in the 135 bp region from –149 to –284 of the ATG site in the human inhibin-α gene promoter, was investigated by bisulfite genomic sequencing (Fig. 1A). Molt-4, Raji, HL-60, and Kasumi-1 cells showed marked hypermethylation of the inhibin-α subunit gene promoter; in contrast, Jurkat cells exhibited hypomethylation. This region was almost unmethylated in IM-9 and K562 cells (Fig. 1B).

Mutations of the inhibin-α gene in human leukemia cells

A mutation study of the inhibin-α gene in human leukemia cells was carried out. The PCR product (fragment A) including nucleotide −16 was digested with SpeI (Fig. 2A). Polymorphisms were identified within the 5′-UTR and exon 1 and used to divide the cell lines into the following two groups: i) CC genotype (Molt-4, Raji, and HL-60 cells) and ii) CT genotype (Jurkat and IM-9 cells) + TT genotype (Kasumi-1 and K562 cells). Interestingly, inhibin-α gene mutation patterns differed between lymphoid leukemia cells (CT, heterozygote) and myeloid leukemia cells (TT, homozygote). Substitution 769G>A of exon 2 in human leukemia cells was analyzed by restriction enzyme digestion. A PCR product comprising nucleotide 769, fragment B, was digested with BsrFI (Fig. 2B) and/or Fnu4HI (Fig. 2C). The single base change at 769G>A of exon 2 was not found in the seven human leukemia cell lines.

LOH at 2q in human leukemia cells

LOH was determined by PCR of genomic DNA. Analysis of the 2q chromosome arm revealed that LOH with at least one microsatellite marker occurred at 2q32-36 in Jurkat, Molt-4, Raji, IM-9, HL-60, Kasumi-1, and K562 cells (Fig. 2D). LOH at 2q32-33 was observed in human lymphoid (Molt-4, Raji, IM-9) and myeloid (K562) leukemia cells. LOH at 2q33-36 was observed in human myeloid (HL-60, Kasumi-1 and K562) leukemia cells, but not in human lymphoid leukemia cells. However, K562 cells exhibited LOH at both 2q32-33 and 2q33-36.

Effect of 5-AzaC treatment on inhibin-α and betaglycan mRNA levels in human leukemia cells

Basal expression of inhibin-α mRNA was not detected in human leukemia cells, whereas betaglycan mRNA was expressed in the majority of cells (Fig. 3A). To evaluate the role of methylation in the inactivation of the inhibin-α gene promoter in human leukemia cells, a DNA methyltransferase inhibitor, 5-AzaC, was used. Human leukemia cells were treated with 2 and 5 μM 5-AzaC, and inhibin-α and betaglycan mRNA levels were measured by real-time PCR (Fig. 3B). 5-AzaC treatment resulted in increased inhibin-α and betaglycan mRNA levels in all seven human leukemia cell lines. The magnitude of the increase in inhibin-α and betaglycan mRNA levels caused by 5-AzaC treatment was greater in lymphoid than in myeloid leukemia cells.

Effect of 5-AzaC treatment on inhibin-α protein levels in human leukemia cells

Human leukemia cells were treated with 5 μM 5-AzaC, and inhibin-α subunit protein levels were measured by flow cytometry and FITC staining (Fig. 4). Treatment with 5-AzaC resulted in 11.3–32.3- and 14.0–27.2- fold increases in inhibin-α protein levels in human lymphoid and myeloid leukemia cells. Fluorescence intensities after 5-AzaC treatment were higher in human lymphoid compared to myeloid leukemia cells.

Effect of 5-AzaC on the growth and doubling time of human leukemia cells

Cells exposed to 0.5, 2, and 5 μM 5-AzaC exhibited significant growth inhibition in a dose-dependent manner, and the population doubling time of human leukemia cells was increased by 1.3–2.6-fold (Table I).

Table I

Suppression by 5-AzaC of the growth of human leukemia cells.

Table I

Suppression by 5-AzaC of the growth of human leukemia cells.

Human leukemia cell linesViability (%)
Doubling time (h)
Fold growth suppression
0 μM5-AzaC
5.0 μM5-AzaC
0.5 μM2.0 μM0 μM5.0 μM
Jurkat10028.828.828.120502.5
Molt-410042.140.639.536681.9
Raji10024.923.221.926572.2
IM-910038.635.633.234551.6
HL-6010082.473.666.527692.6
Kasumi-110076.571.167.767831.3
K56210088.282.881.147841.8

[i] Cells (5×105/100 mm dish) were treated with 5-AzaC (0, 0.5, 2.0 and 5.0 μM) for 5 days. Viable cells were enumerated by trypan blue assay. At the end of the treatment period, the cells were seeded in 12-well plates at 5×104/ml. Cell number/plate was determined by trypan blue assay daily for 5 consecutive days. Untreated cells were analyzed under similar conditions as a control. The average cell number from two plates was determined, and the mean cell numbers were plotted to calculatethe cell population doubling times.

Effect of 5-AzaC on the cell cycle in human leukemia cells

The cell cycle profiles of human leukemia cells treated with 5 μM 5-AzaC were analyzed by flow cytometry (Fig. 5). The results suggested changes in the cell cycle and induction of apoptosis in 5-AzaC-treated cells. Treatment of Jurkat, Molt-4, Raji, IM-9, HL-60, Kasumi-1, and K562 cells with 5-AzaC resulted in a 1.7–36.2-fold increase in the proportion of apoptotic cells. Interestingly, human lymphoid leukemia cells exhibited a greater increase in the proportion of apoptotic cells than myeloid leukemia cells after treatment with 5-AzaC.

Discussion

Methylation of CpG sites within the regulatory regions of tumor-suppressor genes is a common aberration in human cancers that is frequently associated with gene silencing. In this study, the degree of methylation varied among the seven CpG sites in the inhibin-α gene promoter in four human lymphoid, and three human myeloid leukemia cells. Seven CpG sites were significantly hypermethylated in human lymphoid (Molt-4 and Raji) leukemia cells and human myeloid (HL-60 and Kasumi-1) leukemia cells, while lymphoid (Jurkat) leukemia cells exhibited hypomethylation. In contrast, this region was not methylated in lymphoid (IM-9) or myeloid (K562) leukemia cells. The inhibin-α subunit PC3 prostate cancer cell lines. The methylation pattern ranges from dense to sparse methylation, with CpG sites 0–3 being undermethylated in the DU145 and PC3 cell lines compared to the LNCaP cells (20). Our findings suggested that the methylation pattern at CpG sites did not differ significantly between human lymphoid and myeloid leukemia cells. Germline cells of chronic lymphocytic leukemia (CLL) patients with allele-specific expression (ASE) showed increased levels of DNA methylation in the promoter region (26).

Transcriptional silencing of tumor-suppressor genes can be caused by mutations. Two polymorphic sites were identified: −16>T in the 5-UTR and 769G>A in exon 2. Mutation at the −16-bp site of the 5′-UTR was heterozygotic in lymphoid (Jurkat and IM-9) cells and homozygotic in myeloid (K562) cells. The −16-bp mutation in the 5′-UTR differed significantly between human lymphoid and myeloid leukemia cells, suggesting that human leukemia cells may be affected by the −16>T allele variant and supporting the concept that the 5′-UTR allele variant. In this study, Molt-4, Raji, IM-9, HL-60, and Kasumi-1 cells showed LOH at chromosome 2q32-36, but Jurkat cells did not. In contrast, K562 cells exhibited LOH at both 2q32-36 and 2q33-36. Changes at chromosome 2q occur in prostate carcinoma and pediatric adrenocortical tumors (2729). In bladder carcinoma and head-and-neck squamous cell carcinoma (30,31), 2q deletion is correlated with advanced disease and a poor prognosis. Taken together, our results suggest that low expression of the inhibin-α subunit gene is related to hypermethylation, mutation and LOH.

Induction of inhibin-α subunit mRNA expression in the human gastric cancer cell lines by treatment with 5-AzaC demonstrates the presence of all necessary transcription factors (19). In the human leukemia cell lines analyzed in this study, expression of inhibin-α subunit mRNA after 5-AzaC treatment was not correlated with methylation status. In prostate cancer cell lines, expression of inhibin-α subunit mRNA was correlated with methylation status after treatment with 5-AzaC and trichostatin A (TSA). A reciprocal relationship between the degree of methylation and re-expression of inhibin-α subunit was evident after treatment with 5-AzaC. PC3 cells, which exhibited the lowest degree of methylation, were easily demethylated and expressed high levels of inhibin-α subunit mRNA; in contrast, LNCaP cells, which were the most highly methylated, showed lower expression of inhibin-α subunit mRNA (20). These results suggest that the expression level is not dependent on the degree of methylation within the promoter region.

The pattern of methylation of the inhibin-α gene reflected the level of the encoded protein in human leukemia cells. Immunostaining of 5-AzaC-treated-cells was performed to evaluate inhibin-α protein levels. Human myeloid leukemia (HL-60 and Kasumi-1) cells treated with 5-AzaC showed lower inhibin-α protein levels than the other cell lines. Interestingly, the increase in inhibin-α protein levels was greater in human lymphoid than in myeloid leukemia cells. However, the inhibin-α subunit protein level was not correlated with the methylation status of those cell lines after 5-AzaC treatment. The percentage of positively stained demethylated LNCaP and DU145 cells was lower compared with that of demethylated PC3 cells (20). Sequential gene expression changed in cancer cell lines after treatment with 5-AzaC and then focused on the genes with expression levels that changed gradually, because the effect of hypomethylation by 5-AzaC would gradually occur. Monitoring of changes in mRNA levels after 5-AzaC treatment enables identification of genes whose expression levels changed gradually (32). 5-AzaC is a DNA demethylating and anti-cancer agent resulting in the induction of genes suppressed via DNA hypermethylation (33). Some of the genes upregulated by 5-AzaC treatment may be transcriptionally repressed by promoter hypermethylation in gastric cancer (34). We found a correlation between inhibin-α mRNA and protein levels in human leukemia cells after treatment with 5-AzaC. Also, betaglycan mRNA levels were influenced by 5-AzaC treatment. However, pattern of increases of inhibin-α and betaglycan mRNA showed correlation in gene expression between human lymphoid and myeloid leukemia cells.

Our data show that treatment with 5-AzaC led to a substantial increase in the doubling times of surviving leukemia cells, and an increased proportion of apoptotic cells due to nonspecific suppression of cell growth. 5-AzaC-induced growth inhibition results from the release of methylation silencing of cell cycle regulatory genes, such as p16 (35). Moreover, 5-AzaC affects the levels of several proteins involved in cell cycle regulation, apoptosis, and survival (36,37). Our results suggest that inhibin-α has a critical function in cells. 5-AzaC exerts a cytotoxic effect.

In this study, epigenetically mediated aberrant transcriptional silencing of the inhibin-α gene in human leukemia cells was characterized. The results suggested that this gene likely plays an important role in leukemia tumorigenesis as a putative tumor suppressor. Methylation of the inhibin-α gene promoter was evident in some human leukemia cell lines, but whether this is a cause or consequence of tumorigenesis remains to be determined. Moreover, functional studies of the inhibin-α gene may provide insight into the development of leukemia treatment. Our results suggest that the inhibin-α gene promoter is poised for activation in the cell lines tested and that the effects on transcription are primarily indirect and mediated by activation of transcription factors. Induction of the expression of other genes, either alone or in combination with inhibin-α, may explain the observed growth suppression in human leukemia cells.

References

1 

Antequera F, Boyes J and Bird A: High levels of de novo methylation and altered chromatin structure at CpG islands in cell lines. Cell. 62:503–514. 1990. View Article : Google Scholar : PubMed/NCBI

2 

Esteller M: CpG island hypermethylation and tumor suppressor genes: A booming present, a brighter future. Oncogene. 21:5427–5440. 2002. View Article : Google Scholar : PubMed/NCBI

3 

Okano M, Xie S and Li E: Cloning and characterization of a family of novel mammalian DNA (cytosine-5) methyltransferases. Nat Genet. 19:219–220. 1998. View Article : Google Scholar : PubMed/NCBI

4 

Jones PL, Veenstra GJ, Wade PA, Vermaak D, Kass SU, Landsberger N, Strouboulis J and Wolffe AP: Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription. Nat Genet. 19:187–191. 1998. View Article : Google Scholar : PubMed/NCBI

5 

Nan X, Ng HH, Johnson CA, Laherty CD, Turner BM, Eisenman RN and Bird A: Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature. 393:386–389. 1998. View Article : Google Scholar : PubMed/NCBI

6 

van der Ploeg LH and Flavell RA: DNA methylation in the human gamma delta beta-globin locus in erythroid and nonerythroid tissues. Cell. 19:947–958. 1980. View Article : Google Scholar : PubMed/NCBI

7 

Lichtenstein M, Keini G, Cedar H and Bergman Y: B cell-specific demethylation: A novel role for the intronic κ chain enhancer sequence. Cell. 76:913–923. 1994. View Article : Google Scholar : PubMed/NCBI

8 

Lübbert M, Miller CW and Koef fler HP: Changes of DNA methylation and chromatin structure in the human myeloperoxidase gene during myeloid differentiation. Blood. 78:345–356. 1991.PubMed/NCBI

9 

Felgner J, Kreipe H, Heidorn K, Jaquet K, Heuss R, Zschunke F, Radzun HJ and Parwaresch MR: Lineage-specific methylation of the c-fms gene in blood cells and macrophages. Leukemia. 6:420–425. 1992.PubMed/NCBI

10 

Baylin SB, Herman JG, Graff JR, Vertino PM and Issa JP: Alterations in DNA methylation: A fundamental aspect of neoplasia. Adv Cancer Res. 72:141–196. 1998. View Article : Google Scholar

11 

Toyooka S, Carbone M, Toyooka KO, Bocchetta M, Shivapurkar N, Minna JD and Gazdar AF: Progressive aberrant methylation of the RASSF1A gene in simian virus 40 infected human mesothelial cells. Oncogene. 21:4340–4344. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Tsai CN, Tsai CL, Tse KP, Chang HY and Chang YS: The Epstein- Barr virus oncogene product, latent membrane protein 1, induces the downregulation of E-cadherin gene expression via activation of DNA methyltransferases. Proc Natl Acad Sci USA. 99:10084–10089. 2002. View Article : Google Scholar

13 

Mathews LS: Activin receptors and cellular signaling by the receptor serine kinase family. Endocr Rev. 15:310–325. 1994. View Article : Google Scholar : PubMed/NCBI

14 

Mather JP, Moore A and Li RH: Activins, inhibins, and follistatins: Further thoughts on a growing family of regulators. Proc Soc Exp Biol Med. 215:209–222. 1997. View Article : Google Scholar : PubMed/NCBI

15 

Risbridger GP, Schmitt JF and Robertson DM: Activins and inhibins in endocrine and other tumors. Endocr Rev. 22:836–858. 2001. View Article : Google Scholar : PubMed/NCBI

16 

Matzuk MM, Finegold MJ, Su JG, Hsueh AJ and Bradley A: α-inhibin is a tumour-suppressor gene with gonadal specificity in mice. Nature. 360:313–319. 1992. View Article : Google Scholar : PubMed/NCBI

17 

Matzuk MM and Bradley A: Identification and analysis of tumor suppressor genes using transgenic mouse models. Semin Cancer Biol. 5:37–45. 1994.PubMed/NCBI

18 

Schmitt JF, Millar DS, Pedersen JS, Clark SL, Venter DJ, Frydenberg M, Molloy PL and Risbridger GP: Hypermethylation of the inhibin α-subunit gene in prostate carcinoma. Mol Endocrinol. 16:213–220. 2002.PubMed/NCBI

19 

Kim YI, Shim J, Kim BH, Lee SJ, Lee HK, Cho C and Cho BN: Transcriptional silencing of the inhibin-α gene in human gastric carcinoma cells. Int J Oncol. 41:690–700. 2012.PubMed/NCBI

20 

Balanathan P, Ball EM, Wang H, Harris SE, Shelling AN and Risbridger GP: Epigenetic regulation of inhibin α-subunit gene in prostate cancer cell lines. J Mol Endocrinol. 32:55–67. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Chatterton Z, Burke D, Emslie KR, Craig JM, Ng J, Ashley DM, Mechinaud F, Saffery R and Wong NC: Validation of DNA methylation biomarkers for diagnosis of acute lymphoblastic leukemia. Clin Chem. 60:995–1003. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Liu P, Shen JK, Xu J, Trahan CA, Hornicek FJ and Duan Z: Aberrant DNA methylations in chondrosarcoma. Epigenomics. 8:1519–1525. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Clark SJ, Harrison J, Paul CL and Frommer M: High sensitivity mapping of methylated cytosines. Nucleic Acids Res. 22:2990–2997. 1994. View Article : Google Scholar : PubMed/NCBI

24 

Sundblad V, Chiauzzi VA, Andreone L, Campo S, Charreau EH and Dain L: Controversial role of inhibin α-subunit gene in the aetiology of premature ovarian failure. Hum Reprod. 21:1154–1160. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Kuchler RJ: Development of animal cell populations in vitro. Biochemical Methods in Cell Culture and Virology. Kuchler RJ: Dowden, Hutchinson and Ross Inc. Press; Stroudsburg, PA: pp. 90–113. 1977

26 

Wei QX, Claus R, Hielscher T, Mertens D, Raval A, Oakes CC, Tanner SM, de la Chapelle A, Byrd JC, Stilgenbauer S, et al: Germline allele-specific expression of DAPK1 in chronic lymphocytic leukemia. PLoS One. 8:e552612013. View Article : Google Scholar : PubMed/NCBI

27 

Alers JC, Rochat J, Krijtenburg PJ, Hop WC, Kranse R, Rosenberg C, Tanke HJ, Schröder FH and van Dekken H: Identification of genetic markers for prostatic cancer progression. Lab Invest. 80:931–942. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Suarez BK, Lin J, Burmester JK, Broman KW, Weber JL, Banerjee TK, Goddard KA, Witte JS, Elston RC and Catalona WJ: A genome screen of multiplex sibships with prostate cancer. Am J Hum Genet. 66:933–944. 2000. View Article : Google Scholar : PubMed/NCBI

29 

Longui CA, Lemos-Marini SH, Figueiredo B, Mendonca BB, Castro M, Liberatore R Jr, Watanabe C, Lancellotti CL, Rocha MN, Melo MB, et al: Inhibin α-subunit (INHA) gene and locus changes in paediatric adrenocortical tumours from TP53 R337H mutation heterozygote carriers. J Med Genet. 41:354–359. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Zhao J, Richter J, Wagner U, Roth B, Schraml P, Zellweger T, Ackermann D, Schmid U, Moch H, Mihatsch MJ, et al: Chromosomal imbalances in noninvasive papillary bladder neoplasms (pTa). Cancer Res. 59:4658–4661. 1999.PubMed/NCBI

31 

Ransom DT, Barnett TC, Bot J, de Boer B, Metcalf C, Davidson JA and Turbett GR: Loss of heterozygosity on chromosome 2q: Possibly a poor prognostic factor in head and neck cancer. Head Neck. 20:404–410. 1998. View Article : Google Scholar : PubMed/NCBI

32 

Arai M, Yokosuka O, Hirasawa Y, Fukai K, Chiba T, Imazeki F, Kanda T, Yatomi M, Takiguchi Y, Seki N, et al: Sequential gene expression changes in cancer cell lines after treatment with the demethylation agent 5-Aza-2′-deoxycytidine. Cancer. 106:2514–2525. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Sayar N, Karahan G, Konu O, Bozkurt B, Bozdogan O and Yulug IG: Transgelin gene is frequently downregulated by promoter DNA hypermethylation in breast cancer. Clin Epigenetics. 7:1042015. View Article : Google Scholar : PubMed/NCBI

34 

Mikata R, Yokosuka O, Fukai K, Imazeki F, Arai M, Tada M, Kurihara T, Zhang K, Kanda T and Saisho H: Analysis of genes upregulated by the demethylating agent 5-aza-2′-deoxycytidine in gastric cancer cell lines. Int J Cancer. 119:1616–1622. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Bender CM, Pao MM and Jones PA: Inhibition of DNA methylation by 5-aza-2′-deoxycytidine suppresses the growth of human tumor cell lines. Cancer Res. 58:95–101. 1998.PubMed/NCBI

36 

Song SH, Jong HS, Choi HH, Inoue H, Tanabe T, Kim NK and Bang YJ: Transcriptional silencing of Cyclooxygenase-2 by hyper-methylation of the 5′ CpG island in human gastric carcinoma cells. Cancer Res. 61:4628–4635. 2001.PubMed/NCBI

37 

Valdez BC, Li Y, Murray D, Corn P, Champlin RE and Andersson BS: 5-Aza-2′-deoxycytidine sensitizes busulfan- resistant myeloid leukemia cells by regulating expression of genes involved in cell cycle checkpoint and apoptosis. Leuk Res. 34:364–372. 2010. View Article : Google Scholar

Related Articles

Journal Cover

April-2017
Volume 50 Issue 4

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kim YI, Park S, Kwon HS, Yang H, Cho SY, Kim YJ and Lee HJ: Inhibin-α gene mutations and mRNA levels in human lymphoid and myeloid leukemia cells. Int J Oncol 50: 1403-1412, 2017
APA
Kim, Y.I., Park, S., Kwon, H.S., Yang, H., Cho, S.Y., Kim, Y.J., & Lee, H.J. (2017). Inhibin-α gene mutations and mRNA levels in human lymphoid and myeloid leukemia cells. International Journal of Oncology, 50, 1403-1412. https://doi.org/10.3892/ijo.2017.3895
MLA
Kim, Y. I., Park, S., Kwon, H. S., Yang, H., Cho, S. Y., Kim, Y. J., Lee, H. J."Inhibin-α gene mutations and mRNA levels in human lymphoid and myeloid leukemia cells". International Journal of Oncology 50.4 (2017): 1403-1412.
Chicago
Kim, Y. I., Park, S., Kwon, H. S., Yang, H., Cho, S. Y., Kim, Y. J., Lee, H. J."Inhibin-α gene mutations and mRNA levels in human lymphoid and myeloid leukemia cells". International Journal of Oncology 50, no. 4 (2017): 1403-1412. https://doi.org/10.3892/ijo.2017.3895