Abstract
Aim:
The current study investigated the effect of trichostatin A (TSA) on mitochondrial/intrinsic [pro- (Bax, Bak, and Bim) and anti- (Bcl-2, Bcl-xL, and Mcl-1) apoptotic genes] and cytoplasmic/extrinsic (DR4, DR5, FAS, FAS-L, and TRAIL genes) pathways, histone deacetylase 1, 2, and 3, p53, p73, cell viability, and apoptosis in hepatocellular carcinoma (HCC) HCCLM3, MHCC97H, and MHCC97L cell lines.
Background:
Modulation of the acetylation status of histones, histones modification, plays an important role in regulating gene transcription and expression. Histone deacetylation controlled by histone deacetylases (HDACs) leads to gene downregulation. Histone deacetylase inhibitors (HDACIs) are an emerging class of therapeutics with potential anticancer effects. They can induce apoptosis by activating both extrinsic and intrinsic apoptotic pathways
Methods:
HCCLM3, MHCC97H, and MHCC97L cells were cultured and treated with TSA. To determine viability, apoptosis, and the relative expression level of the mentioned genes, MTT assay, cell apoptosis assay, and qRT-PCR, respectively, were conducted.
Results:
TSA up-regulated Bax, Bak, Bim, DR4, DR5, FAS, FAS-L, TRAIL, p53, and p73 and down-regulated Bcl-2, Bcl-xL, Mcl-1, histone deacetylases 1, 2, and 3 significantly, resulting in apoptosis induction. Maximal and minimal apoptosis was seen in the MHCC97H and HCCLM3 cell lines (93.94% and 39.68%, respectively) after 24 and 48 h. Therefore, the MHCC97H cell line was more sensitive to TSA.
Conclusion:
The current findings demonstrated that the HDAC inhibitor TSA can induce apoptosis and inhibit cell growth through both mitochondrial/intrinsic and cytoplasmic/extrinsic apoptotic pathways in hepatocellular carcinoma HCCLM3, MHCC97H, and MHCC97L cell lines.
Key Words: Trichostatin A, Extrinsic, Intrinsic, Pathway, Apoptosis
Introduction
Modulation of the acetylation status of histones, histones modification, plays an important role in regulating gene transcription and expression. Increased histone acetylation increases gene transcription, whereas histone deacetylation induces gene silencing leading to gene downregulation. Histone acetylation is controlled by the opposing actions of two groups of enzymes, i.e. histone acetyltransferases (HATs) and histone deacetylases (HDACs). These two groups determine the pattern and status of the histone acetylation. The activity of HDACs has been linked to tumorigenesis (1). There are two mechanisms by which histone acetylation increases transcriptional activity. HATs transfer the acetyl moiety of acetyl coenzyme A by which the positive charge of the histone tails is neutralized and the structure of the chromatin is relaxed. This structure enables the transcriptional machinery to access the DNA and enhances gene transcription. Just the opposite, HDACs remove the acetyl group from the histone tails, leading to chromatin compaction and decreased gene transcription (2). In humans, there are 18 HDACs divided into three classes based on homology to yeast HDACs (3).
Histone deacetylase inhibitors (HDACIs) are an emerging class of therapeutic compounds with potential anticancer effects. They interfere with HDAC activity resulting in the regulation of biological events, such as cell differentiation, cell cycle, and apoptosis in cancer cells. These compounds bind to and inhibit HDAC enzymatic activity. To date, more than 50 naturally occurring or synthetic HDACIs have been developed. The biochemical structures of these drugs are extremely heterogeneous, from simple agents like valproate to more complicated compounds, such as MS-275. Crystallographic studies have indicated that the hydroxamic acid-based HDACIs suberoylanilide hydroxamic acid (SAHA) and trichostatin A (TSA) fit very well into the HDAC catalytic pocket (4). TSA was one of the first natural hydroxamate compounds found to inhibit HDAC activity, resulting in the reactivation of silenced tumor suppressor genes (TSGs) and apoptosis induction (5).
It has been reported that cell apoptosis is mediated by way of two molecular pathways leading to caspase activation, i.e. extrinsic death receptor and intrinsic mitochondrial pathways. The extrinsic pathway is initiated by ligations of transmembrane death receptors, whereas the intrinsic pathway requires disruption of the mitochondrial membrane to release cytochrome c. The B cell lymphoma-2 (BCL-2) family of proteins regulates the release of mitochondrial cytochrome c: BCL-2 and BCL-XL prevent the release of cytochrome c, whereas BAX promotes the release of cytochrome C (6). The BCL-2 family of proteins is the key regulator of apoptosis, or programmed cell death. They are located predominantly on the mitochondria, where the release of cytochrome c is regulated. Structurally, this family is divided into two major sub-groups, the anti-apoptotic proteins (e.g., BCL-2, BCL-XL, MCL-1, BFL-1, BCL-W, and BCL2L10) and the pro-apoptotic proteins (such as BAK, BAX, BOK, BIM, PUMA, NOXA, BID, etc.) (7).
The extrinsic pathway is triggered by the binding of death ligands of the tumor necrosis factor (TNF) family, such as TNF-related apoptosis-inducing ligand (TRAIL or Apo2L), to their appropriate death receptors (DRs) on the cell surface (8). TRAIL is a type II transmembrane protein, which was originally identified based on sequence homology to Fas ligand (FasL) and TNF. It exerts its function by engaging its receptors expressed on the surface of target cells. To date, four human receptors specific for TRAIL have been reported, comprising TRAIL-R1/DR4, TRAIL-R2/DR5, TRAIL-R3/DcR1, and TRAIL-R4/DcR2 (9). As mentioned, the extrinsic pathway is initiated by the binding of death receptors, including tumor necrosis factor (TNF) receptor-1 (TNFR-1), Fas (Apo-1 or CD95), TNF-related apoptosis-inducing ligand (TRAIL orApo2-L) receptors (DR-4 and -5), and DR-3 (Apo3) and DR-6, to their ligands, such as TNF, FasL, TRAIL, and TL1A (Apo3L) (10, 11).
HDACIs have been shown to upregulate the expression of both death receptors and their ligands in transformed cells (12, 13). In vitro studies have demonstrated that HDACIs induce apoptosis through activation of both extrinsic and intrinsic apoptotic pathways (14). It has been indicated that the histone deacetylase inhibitor TSA induces Bax-dependent apoptosis in colorectal cancer cell lines by both p53-dependent mechanisms (15). Moreover, p53 is essential for the apoptotic response to TSA and SAHA in ovarian cancer, which is associated with activation of caspase-9, caspase-2, caspase-8, and caspase-7 (16). Other researchers have shown that TSA induces apoptosis by restoring both p73 and Bax but not p53 expression (17). Additionally, it has been reported that TSA induces the cell apoptosis of NCI-H157 human lung cancer cells through a signaling cascade of Fas/FasL-mediated extrinsic and mitochondrial-mediated intrinsic caspases pathway (18).
The current study investigated the effects of TSA on mitochondrial/intrinsic [pro- (Bax, Bak, and Bim) and anti- (Bcl-2, Bcl-xL, and Mcl-1) apoptotic genes] and cytoplasmic/extrinsic (DR4, DR5, FAS, FAS-L, and TRAIL genes) pathways, histone deacetylase 1, 2, and 3, p53, p73, cell viability, and apoptosis in hepatocellular carcinoma (HCC) HCCLM3, MHCC97H, and MHCC97L cell lines.
Materials
Materials
Hepatocellular carcinoma HCCLM3, MHCC97H, and MHCC97L cell lines were purchased from the National Cell Bank of Iran-Pasteur Institute. TSA and Dulbecco’s modified Eagle’s medium (DMEM) were obtained from Sigma (St. Louis, MO, USA) and dissolved in dimethyl sulfoxide (DMSO) to make a working stock solution. Further concentrations of TSA were obtained by diluting the provided stock solution. Other necessary materials and kits were purchased as provided for previous works (19, 20). The HCCLM3, MHCC97H, and MHCC97L cells were maintained in DMEM supplemented with fetal bovine serum 10% and antibiotics in a humidified atmosphere of 5% CO2 in air at 37 ℃. This work is a lab trial study that was approved by the Ethics Committee of Jahrom University of Medical Science with the code number IR.JUMS.REC.1399.053.
Cell culture and cell viability
HCCLM3, MHCC97H, and MHCC97L cells were cultured in DMEM supplemented with 10% FBS and antibiotics (100 U/mL streptomycin and 100 U/mL penicillin) at 37 °C in 5% CO2 for 24 h and then seeded into 96-well plates (3 × 105 cells per well). After 24 h, the medium was replaced with a medium containing TSA with various doses (0, 0.5, 1, 2.5, 5, and 10 μM). The control groups were treated with DMSO at a concentration of 0.05%. After 24 and 48 h, the cells, treated and untreated, were investigated by MTT assay according to standard protocols to determine cell viability. The MTT solution was added to each well for 4 h at 37 ℃, and then the MTT solution was changed by DMSO and shaken for 10 min to dissolve all of the crystals. The MTT assay, a quantitative colorimetric assay, is based on the living cell’s ability to reduce the tetrazolium salt MTT. The mitochondrial succinate-dehydrogenases of viable cells cleave the tetrazolium ring in active mitochondria into formazan crystals which can be dissolved in DMSO. Finally, the optical density was detected by a microplate reader at a wavelength of 570 nM. Each experiment was repeated three times.
Cell apoptosis assay
To determine HCCLM3, MHCC97H, and MHCC97L cell apoptosis, the cells were cultured at a density of 3 × 105 cells/well and treated with TSA, based on the IC50 values indicated in Table 1, for 24 and 48 h. Then, both treated and untreated cells were harvested by trypsinization, washed with cold PBS, and resuspended in binding buffer (1x). Finally, Annexin-V-(FITC) and PI were used according to the protocol to determine the apoptotic cells by FACScan flow cytometry (Becton Dickinson, Heidelberg, Germany).
Table 1.
IC50 values of TSA determined by MTT assay
| Cell line | Duration/Hour | IC50/ μM | LogIC50 | R squared |
|---|---|---|---|---|
| HCCLM3 | 24 | 3.273 | 0.5149 | 0.6560 |
| HCCLM3 | 48 | 1.552 | 0.1908 | 0.9773 |
| MHCC97H | 24 | 2.589 | 0.4131 | 0.9024 |
| MHCC97H | 48 | 1.1908 | 0.2805 | 0.8181 |
| MHCC97L | 24 | 3.622 | 0.5589 | 0.8281 |
| MHCC97L | 48 | 1.908 | 0.2805 | 0.9821 |
Real-time Quantitative Reverse Transcription Polymerase Chain Reaction (qRT-PCR)
To determine the relative expression levels of Bax, Bak, Bim, Bcl-2, Bcl-xL, Mcl-1, DR4, DR5, FAS, FAS-L, TRAIL, histone deacetylase inhibitors 1, 2, and 3, p53, and p73, qRT-PCR analyses were done. The HCCLM3, MHCC97H, and MHCC97L cells (at a density of 3 × 105 cells/well) were treated with TSA, based on the IC50 values indicated in Table 1, for 24 and 48 h, except the control groups which were treated with DMSO only. Then qRT-PCR was done as in our previous works (21). The primer sequences are shown in Table 2 (22-33).
Table 2.
Primer sequences of Bax, Bak, Bim, Bcl-2, Bcl-xL, Mcl-1, DR4, DR5, FAS, FAS-L, TRAIL, histone deacetylase inhibitors 1, 2, and 3, p53, and p73 genes
| Primer | Primer sequences (5' to 3') | Product length | Reference |
|---|---|---|---|
| Bax Forward Reverse |
AGTAACATGGAGCTGCAGAGGAT GCTGCCACTCGGAAAAAGAC | 77 bp | |
| Bak Forward Reverse |
TGAAAAATGGCTTCGGGGCAAG CTCTCAAACGGCTGGTGGCAATC |
367 bp | |
| Bim Forward Reverse |
ATTACCAAGCAGCCGAAGAC TCCGCAAAGAACCTGTCAAT |
101 bp | |
| Bcl-2 Forward Reverse |
TGGCCAGGGTCAGAGTTAAA TGGCCTCTCTTGCGGAGTA | 147 bp | |
| Bcl-xL Forward Reverse |
TCCTTGTCTACGCTTTCCACG GGTCGCATTGTGGCCTTT | 62 bp | |
| Mcl-1 Forward Reverse |
AAAGCCTGTCTGCCAAAT CCTATAAACCCACCACTC |
198 bp | |
| DR4 Forward Reverse |
CAGAACATCCTGGAGCCTGTAAC ATGTCCATTGCCTGATTCTTTGTG | 299 bp | |
| DR5 Forward Reverse |
TGCAGCCGTAGTCTTGATTG GCACCAAGTCTGCAAAGTCA |
389 bp | |
| FAS Forward Reverse |
ATGCTGGGCATCTGGACCCT GCCATGTCCTTCATCACACAA | 366 bp | |
| FAS-L Forward Reverse |
TGGAATTGTCCTGCTTTCTGG TGTTGCAAGATTGACCCCG |
113 bp | |
| TRAIL Forward Reverse |
ACCAACGAGCTGAAGCAGAT TCCTTGATGATTCCCAGGAG |
213 bp | |
| HDAC1 Forward Reverse |
AACCTGCCTATGCTGATGCT CAGGCAATTCGTTTGTCAGA | 374 bp | |
| HDAC2 Forward Reverse |
GGGAATACTTTCCTGGCACA ACGGATTGTGTAGCCACCTC | 314 bp | |
| HDAC3 Forward Reverse |
TGGCTTCTGCTATGTCAACG GCACGTGGGTTGGTAGAAGT | 328 bp | |
| P53 Forward Reverse |
CAGCCAAGTCTGTGACTTGCACGTAC CTATGTCGAAAAGTGTTTCTGTCATC |
292 bp | |
| P73 Forward Reverse |
AACGCTGCCCCAACCACGAG GCCGGTTCATGCCCCCTACA |
231 bp | |
| GAPDH Forward Reverse |
GAAGGTGAAGGTCGGAGTC GAAGATGGTGATGGGATTTC | 172 bp |
Statistical analysis
The database was set up with the SPSS 16.0 software package (SPSS Inc., Chicago, Illinois, USA) and Graph Pad Prism 8.0 for data analysis. Results are expressed as mean ± standard deviation (SD) for n=3 independent experiments. Statistical comparisons between groups were performed with ANOVA (oneway ANOVA) and the Tukey test. A p-value < 0.05 was considered as a significant difference.
Results
Results of cell viability by the MTT assay
The viability of the HCCLM3, MHCC97H, and MHCC97L cells treated with various doses of TSA (0, 0.5, 1, 2.5, 5, and 10 μM) was investigated by MTT assay. As shown in Figure 1, TSA induced significant cell growth inhibition in a dose-dependent manner (p< 0.001). The IC50 value was calculated by Graph pad prism 8 as indicated in Table 1.
Figure 1.
Effect of TSA on viability of HCCLM3, MHCC97H, and MHCC97L cells. The cells were treated with and without TSA, and cell viability was evaluated by MTT assay. Each experiment was conducted in triplicate. Mean values from the three experiments ± standard error of mean are shown. Asterisks indicate significant differences between treated and untreated cells
Results of determination of cell apoptosis
To determine cell apoptosis, HCCLM3, MHCC97H, and MHCC97L cells were treated with TSA, based on IC50 values, for 24 and 48 h and then stained using annexin-V-(FITC) and PI to determine apoptotic cells in the early and late stages of apoptosis. As indicated in Figures 2-6, TSA induced significant cell apoptosis in all three cell lines (p<0.001).
Figure 2.
The apoptosis-inducing effect of TSA was investigated by flow cytometric analysis of HCCLM3 cells stained with Annexin V and propidium iodide. The results indicated that TSA significantly induced cell apoptosis after 24 and 48 h of treatment
Figure 6.
Comparative analysis of the effects of TSA on HCCLM3, MHCC97H, and MHCC97L cells (A). Maximal and minimal apoptosis was seen in the MHCC97H and HCCLM3 cell lines, respectively, after 24 and 48 h (B).
Figure 3.
The apoptosis-inducing effect of TSA was investigated by flow cytometric analysis of MHCC97H cells stained with Annexin V and propidium iodide. The results indicated that TSA significantly induced cell apoptosis after 24 and 48 h of treatment
Figure 4.
The apoptosis-inducing effect of TSA was investigated by flow cytometric analysis of MHCC97L cells stained with Annexin V and propidium iodide. The results indicated that TSA significantly induced cell apoptosis after 24 and 48 h of treatment
Results of determining gene expression in HCCLM3 cell line
The effects of TSA on Bax, Bak, Bim, Bcl-2, Bcl-xL, Mcl-1, DR4, DR5, FAS, FAS-L, TRAIL, histone deacetylases 1, 2, and 3, p53, and p73 were evaluated by quantitative real-time RT-PCR analysis. The result of quantitative reverse transcription-polymerase chain reaction analysis demonstrated that this compound up-regulated Bim, DR4, DR5, FAS, FAS-L, TRAIL, p53, and p73 and down-regulated Mcl-1, histone deacetylases 1, 2, and 3 significantly after 24 h of treatment. It had no significant effect on the expression of Bax, Bak, Bcl-2, or Bcl-xL after 24 h. Furthermore, this compound up-regulated Bax, Bak, Bim, DR4, DR5, FAS, FAS-L, TRAIL, p53, and p73 and down-regulated Bcl-2, Bcl-xL, Mcl-1, and histone deacetylases 1, 2, and 3 significantly after 48 h of treatment (Figure 7).
Figure 7.
Relative expression levels of Bax, Bak, Bim, Bcl-2, Bcl-xL, Mcl-1, DR4, DR5, FAS, FAS-L, TRAIL, histone deacetylase inhibitors 1, 2, and 3, p53, and p73 in HCCLM3 cells treated with TSA for 24 and 48 h. Quantitative reverse transcription-polymerase chain reaction analysis demonstrated that this compound up-regulated Bim, DR4, DR5, FAS, FAS-L, TRAIL, p53, and p73 and down-regulated Mcl-1, histone deacetylases 1, 2, and 3 significantly after 24 h of treatment. It had no significant effect on the expression of Bax, Bak, Bcl-2, and Bcl-xL after 24 h. Furthermore, this compound up-regulated Bax, Bak, Bim, DR4, DR5, FAS, FAS-L, TRAIL, p53, and p73 and down-regulated Bcl-2, Bcl-xL, Mcl-1, and histone deacetylases 1, 2, and 3 significantly after 48 h of treatment. Asterisks indicate significant differences between treated cells and the control group. Data is presented as means ± standard error. ****p<0.0001
Figure 5.
Apoptotic effects of TSA on HCCLM3, MHCC97H, and MHCC97L cells versus control groups at different periods (24 and 48h). Results were obtained from three independent experiments and expressed as mean ± standard error. Statistical analysis indicated significant differences between treated and untreated cells (A2, B2, and C2).
Results of determining gene expression in MHCC97H cell line
The effects of TSA on the Bax, Bak, Bim, Bcl-2, Bcl-xL, Mcl-1, DR4, DR5, FAS, FAS-L, TRAIL, histone deacetylases 1, 2, and 3, p53, and p73 were evaluated by quantitative real-time RT-PCR analysis. Quantitative reverse transcription-polymerase chain reaction analysis demonstrated that this compound up-regulated Bax, Bak, Bim, DR4, DR5, FAS, FAS-L, TRAIL, p53, and p73 and down-regulated Bcl-2, Bcl-xL, Mcl-1, and histone deacetylases 1, 2, and 3 significantly after 24 and 48 h of treatment (Figure 8).
Figure 8.
Relative expression levels of Bax, Bak, Bim, Bcl-2, Bcl-xL, Mcl-1, DR4, DR5, FAS, FAS-L, TRAIL, histone deacetylase inhibitors 1, 2, and 3, p53, and p73 in MHCC97H cells treated with TSA for 24 and 48 h. Quantitative reverse transcription-polymerase chain reaction analysis demonstrated that this compound up-regulated Bax, Bak, Bim, DR4, DR5, FAS, FAS-L, TRAIL, p53, and p73 and down-regulated Bcl-2, Bcl-xL, Mcl-1, and histone deacetylases 1, 2, and 3 significantly after 24 and 48 h of treatment. Asterisks indicate significant differences between treated cells and the control group. Data is presented as means ± standard error. ****p and **p <0.0001 and p <0.0077, respectively
Results of determining gene expression in MHCC97L cell
The effects of TSA on Bax, Bak, Bim, Bcl-2, Bcl-xL, Mcl-1, DR4, DR5, FAS, FAS-L, TRAIL, and histone deacetylases 1, 2, and 3, p53, and p73 were evaluated by quantitative real-time RT-PCR analysis. Quantitative reverse transcription-polymerase chain reaction analysis demonstrated that this compound up-regulated Bim, DR4, DR5, FAS, FAS-L, TRAIL, p53, and p73 and down-regulated Bcl-2, and Bcl-xL, Mcl-1, and histone deacetylases 1, 2, and 3 significantly after 24 h of treatment. It had no significant effect on the expression of Bax and Bak after 24 h. Furthermore, this compound up-regulated Bax, Bak, Bim, DR4, DR5, FAS, FAS-L, TRAIL, p53, and p73 and down-regulated Bcl-2, Bcl-xL, Mcl-1, and histone deacetylases 1, 2, and 3 significantly after 48 h of treatment (Figure 9).
Figure 9.
Relative expression levels of Bax, Bak, Bim, Bcl-2, Bcl-xL, Mcl-1, DR4, DR5, FAS, FAS-L, TRAIL, histone deacetylase inhibitors 1, 2, and 3, p53, and p73 in MHCC97L cells treated with TSA for 24 and 48 h. Quantitative reverse transcription-polymerase chain reaction analysis demonstrated that this compound up-regulated Bim, DR4, DR5, FAS, FAS-L, TRAIL, p53, and p73 and down-regulated Bcl-2, and Bcl-xL, Mcl-1, and histone deacetylases 1, 2, and 3 significantly after 24 h of treatment. It had no significant effect on the expression of Bax and Bak after 24 h. Furthermore, this compound up-regulated Bax, Bak, Bim, DR4, DR5, FAS, FAS-L, TRAIL, p53, and p73 and down-regulated Bcl-2, Bcl-xL, Mcl-1, and histone deacetylases 1, 2, and 3 significantly after 48 h of treatment. Asterisks indicate significant differences between treated cells and the control group. Data is presented as means ± standard error. p <0.0001
Discussion
Histone deacetylase (HDAC) inhibitors can induce apoptosis through both intrinsic and extrinsic pathways (34). It has been reported that they activate the intrinsic pathway through the upregulation of several pro-apoptotic genes such as Bim, Bid, and Bmf. They engage the extrinsic pathway through the upregulation of DR expression, upregulation of ligands such as TRAIL, and reductions in c-FLIP (35). In vitro studies have demonstrated that histone deacetylase inhibitors induce apoptosis through the signaling cascade of Fas/FasL-mediated extrinsic and mitochondrial-mediated intrinsic caspases pathway (36).
The current findings demonstrated that TSA up-regulated Bax, Bak, Bim, DR4, DR5, FAS, FAS-L, TRAIL, p53, and p73 and down-regulated Bcl-2, Bcl-xL, Mcl-1, and histone deacetylases 1, 2, and 3 significantly, resulting in apoptosis induction.
Similarly, our previous work indicated that TSA can induce apoptosis by inhibiting deacetylases 1, 2, and 3 gene expression and up-regulating p21, p27, and p57 in the breast cancer SK-BR-3 cell line (37). In leukemia cell lines (Jurkat, HL60, K562, and U937), TSA has been shown to induce apoptosis through multiple mechanisms, including the up-regulation of DR4, DR5, Bax, Bak, Bim, PUMA, and Noxa, down-regulation of Mcl-1, Bcl-XL, Bcl-2, and cFLIP, the release of mitochondrial proteins (cytochrome c), induction of p21WAF1/CIP1 and p27KIP1, and activation of caspase-3 (38). Additionally, HDACIs can induce apoptosis in chronic lymphocytic leukemia (CLL) through the inactivation of Bcl-2 family members by increases in Noxa and Bim (39). Other researchers have demonstrated that TSA and valproic acid (VPA) increase the pro-apoptotic Bim level and reduce the anti-apoptotic Mcl-1 level in pancreatic cancer (Panc1 and PaCa44) cells (40).
Moreover, HDACIs increase the activation of caspases and Bid and the inactivation of the anti-apoptotic proteins Bcl-x, XIAP, RIP, and survivin, thereby increasing the pro- to anti-apoptotic protein ratio (41). In colorectal cancer (HCT116 and HT29) cell lines, TSA induces the expression of Bax and decreases the expression of Bcl-2 and Bcl-xL (42).
Many in vitro experiments have proven that these compounds influence DR5 (death receptor 5), death receptors TRAIL (TNF-related apoptosis-inducing ligand), TNF (tumor necrosis factor), Fas (TNF superfamily 6), TNF-related ligands Fas-L, TLA1 (transparent leaf area peptide), and LIGHT (TNF superfamily member 14). It can be concluded that in cancer cells exposed to HDACIs, pro-apoptotic genes involved in the intrinsic (BAK, BAX, and APAF1) and/or extrinsic (FAS, FAS-L, TRAIL, DR5, and TNF-α) apoptotic pathways are up-regulated, while anti-apoptotic genes (XIAP, Bcl-2) are downregulated (43).
The results of the current study indicated that TSA up-regulated p53 and p73 expression significantly. Similar to the current results, it has been reported that TSA induces apoptosis by p53 up-regulation and Bcl-2 down-regulation in the HCC HepG2 cell line (44). Inconsistent with these findings, TSA has been shown to induce apoptosis through Bax up-regulation in human gastric cell lines (45). Similarly, TSA enhances the apoptosis of cervical cancer cells through the overexpression of p53 and p73 (46).
In the present study, TSA had no significant effect on the expression of Bax, Bak, Bcl-2, or Bcl-xL in the HCCLM3 cell line, nor on the expression of Bax and Bak in the MHCC97L cell line after 24 h of treatment. No report on the effects of TSA on the expression of Bax, Bak, Bcl-2, and Bcl-xL in HCCLM3, MHCC97H, and MHCC97L cell lines was found in a literature search. It may be possible to change the expression of the mentioned genes with high concentrations of TSA. Therefore, the evaluation of a high dose of TSA on these cell lines is recommended.
The current findings demonstrate that the HDAC inhibitor TSA can induce apoptosis and inhibit cell growth through both mitochondrial/intrinsic and cytoplasmic/extrinsic apoptotic pathways in hepatocellular carcinoma HCCLM3, MHCC97H, and MHCC97L cell lines.
Acknowledgment
This article was supported by the Adjutancy of Research of Jahrom University of Medical Sciences, Iran.
Conflict of interests
The authors declare that they have no conflict of interest.
References
- 1.Gillenwater AM, Zhong M, Lotan R. Histone deacetylase inhibitor suberoylanilide hydroxamic acid induces apoptosis through both mitochondrial and Fas (Cd95) signaling in head and neck squamous carcinoma cells. Mol Cancer Ther. 2007;6:2967. doi: 10.1158/1535-7163.MCT-04-0344. [DOI] [PubMed] [Google Scholar]
- 2.Ververis K, Hiong A, Karagiannis TC, Licciardi PV. Histone deacetylase inhibitors (HDACIs): multitargeted anticancer agents. Biologics. 2013;7:47–53. doi: 10.2147/BTT.S29965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Dokmanovic M, Marks PA. Prospects: histone deacetylase inhibitors. J Cell Biochem. 2005;96:293–304. doi: 10.1002/jcb.20532. [DOI] [PubMed] [Google Scholar]
- 4.Kim TY, Bang YJ, Robertson KD. Histone deacetylase inhibitors for cancer therapy. Epigenetics. 2006;1:15–24. doi: 10.4161/epi.1.1.2644. [DOI] [PubMed] [Google Scholar]
- 5.Mottamal M, Zheng S, Huang TL, Wang G. Histone deacetylase inhibitors in clinical studies as templates for new anticancer agents. Molecules. 2015;20:3898–941. doi: 10.3390/molecules20033898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Doi S, Soda H, Oka M, Tsurutani J, Kitazaki T, Nakamura Y, Fukuda M, Yamada Y, Kamihira S, Kohno S. The histone deacetylase inhibitor FR901228 induces caspase-dependent apoptosis via the mitochondrial pathway in small cell lung cancer cells. Mol Cancer Ther. 2004;3:1397–02. [PubMed] [Google Scholar]
- 7.Opferman JT, Kothari A. Anti-apoptotic BCL-2 family members in development. Cell Death Differ. 2018;25:37–45. doi: 10.1038/cdd.2017.170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Sayers TJ. Targeting the extrinsic apoptosis signaling pathway for cancer therapy. Cancer Immunol Immunother. 2011;60:1173–80. doi: 10.1007/s00262-011-1008-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Yagita H, Takeda K, Hayakawa Y, Smyth MJ, Okumura K. TRAIL and its receptors as targets for cancer therapy. Cancer Sci. 2004;95:777–83. doi: 10.1111/j.1349-7006.2004.tb02181.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Xu W, Parmigiani R, Marks P. Histone deacetylase inhibitors: molecular mechanisms of action. Oncogene. 2007;26:5541–52. doi: 10.1038/sj.onc.1210620. [DOI] [PubMed] [Google Scholar]
- 11.Ashkenazi A. Targeting death and decoy receptors of the tumour-necrosis factor superfamily. Nat Rev Cancer. 2002;2:420–30. doi: 10.1038/nrc821. [DOI] [PubMed] [Google Scholar]
- 12.Nakata S, Yoshida T, Horinaka M, Shiraishi T, Wakada M, Sakai T. Histone deacetylase inhibitors upregulate death receptor 5/TRAIL-R2 and sensitize apoptosis induced by TRAIL/APO2-L in human malignant tumor cells. Oncogene. 2004;23:6261–71. doi: 10.1038/sj.onc.1207830. [DOI] [PubMed] [Google Scholar]
- 13.Insinga A, Monestiroli S, Ronzoni S, Gelmetti V, Marchesi F, Viale A, et al. Inhibitors of histone deacetylases induce tumor-selective apoptosis through activation of the death receptor pathway. Nat Med. 2005;11:71–76. doi: 10.1038/nm1160. [DOI] [PubMed] [Google Scholar]
- 14.Hrabeta J, Stiborova M, Adam V, Kizek R, Eckschlager T. Histone deacetylase inhibitors in cancer therapy. A review. Biomed Papers. 2014;158:161–9. doi: 10.5507/bp.2013.085. [DOI] [PubMed] [Google Scholar]
- 15.Dai L, He G, Zhang K, Guan X, Wang Y, Zhang B. Trichostatin A induces p53dependent endoplasmic reticulum stress in human colon cancer cells. Oncol Lett. 2019;17:660–7. doi: 10.3892/ol.2018.9641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Henderson C, Mizzau M, Paroni G, Maestro R, Schneider C, Brancolini C. Role of caspases, Bid, and p53 in the apoptotic response triggered by histone deacetylase inhibitors trichostatin-A (TSA) and suberoylanilide hydroxamic acid (SAHA) J Biol Chem. 2003;278:12579–89. doi: 10.1074/jbc.M213093200. [DOI] [PubMed] [Google Scholar]
- 17.Muscolini M, Cianfrocca R, Sajeva A, Mozzetti S, Ferrandina G, Costanzo A, et al. Trichostatin A up-regulates p73 and induces Bax-dependent apoptosis in cisplatin-resistant ovarian cancer cells. Mol Cancer Ther. 2008;7:1410–19. doi: 10.1158/1535-7163.MCT-08-0299. [DOI] [PubMed] [Google Scholar]
- 18.Kim HR, Kim EJ, Yang SH, Jeong ET, Park C, Lee JH, et al. Trichostatin A induces apoptosis in lung cancer cells via simultaneous activation of the death receptor-mediated and mitochondrial pathway. Exp Mol Med. 2006;38:616–24. doi: 10.1038/emm.2006.73. [DOI] [PubMed] [Google Scholar]
- 19.Sanaei M, Kavoosi F, Arabloo M. Effect of Curcumin in Comparison with Trichostatin A on the Reactivation of Estrogen Receptor Alpha Gene Expression, Cell Growth Inhibition and Apoptosis Induction in Hepatocellular Carcinoma Hepa 1-6 Cell lLine. Asian Pac J Cancer Prev. 2020;21:1045–50. doi: 10.31557/APJCP.2020.21.4.1045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Sanaei M, Kavoosi F. Effect of curcumin and trichostatin a on the expression of DNA methyltransfrase 1 in hepatocellular carcinoma cell line hepa 1-6. Iranian J Pediatr Hematol Oncol. 2018;8:193–201. [Google Scholar]
- 21.Sanaei M, Kavoosi F, Roustazadeh A, Shahsavani H. In vitro effect of the histone deacetylase inhibitor valproic acid on viability and apoptosis of the PLC/PRF5 human hepatocellular carcinoma cell line. Asian Pac J Cancer Prev. 2018;19:2507–10. doi: 10.22034/APJCP.2018.19.9.2507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Cao XX, Mohuiddin I, Chada S, Mhashilkar AM, Ozvaran MK, McConkey DJ, et al. Adenoviral transfer of mda-7 leads to BAX up-regulation and apoptosis in mesothelioma cells, and is abrogated by over-expression of BCL-XL. Mol Med. 2002;8:869–76. [PMC free article] [PubMed] [Google Scholar]
- 23.Morrison DJ, English MA, Licht JD. WT1 induces apoptosis through transcriptional regulation of the proapoptotic Bcl-2 family member Bak. Cancer Res. 2005;65:8174–82. doi: 10.1158/0008-5472.CAN-04-3657. [DOI] [PubMed] [Google Scholar]
- 24.Zhang XQ, Dong JJ, Cai T, Shen X, Zhou XJ, Liao L. High glucose induces apoptosis via upregulation of Bim expression in proximal tubule epithelial cells. Oncotarget. 2017;8:24119–25. doi: 10.18632/oncotarget.15491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Xu Y, Liu L, Qiu X, Liu Z, Li H, Li Z, et al. CCL21/CCR7 prevents apoptosis via the ERK pathway in human non-small cell lung cancer cells. PLoS One. 2012;7:33262–71. doi: 10.1371/journal.pone.0033262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Zhang YL, Pang LQ, Wu Y, Wang XY, Wang CQ, Fan Y. Significance of Bcl-xL in human colon carcinoma. World J Gastroenterol. 2008;14:3069–75. doi: 10.3748/wjg.14.3069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Wang B, Ni Z, Dai X, Qin L, Li X, Xu L, et al. The Bcl-2/xL inhibitor ABT-263 increases the stability of Mcl-1 mRNA and protein in hepatocellular carcinoma cells. Mol Cancer. 2014;13:98–104. doi: 10.1186/1476-4598-13-98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Nakamoto N, Higuchi H, Kanamori H, Kurita S, Tada S, Takaishi H, et al. Cyclooxygenase-2 inhibitor and interferon-β synergistically induce apoptosis in human hepatoma cells in vitro and in vivo. Int J Oncol. 2006;29:625–35. [PubMed] [Google Scholar]
- 29.Werner J-M, Kuhl S, Stavrinou P, Roehn G, Krischek B, Blau T, et al. Expression of FAS-L differs from primary to relapsed low-grade gliomas and predicts progression-free survival. Anticancer Res. 2017;37:6639–48. doi: 10.21873/anticanres.12121. [DOI] [PubMed] [Google Scholar]
- 30.Zuo C, Qiu X, Liu N, Yang D, Xia M, Liu J, et al. Interferon-α and cyclooxygenase-2 inhibitor cooperatively mediates TRAIL-induced apoptosis in hepatocellular carcinoma. Exp Cell Res. 2015;333:316–26. doi: 10.1016/j.yexcr.2015.02.013. [DOI] [PubMed] [Google Scholar]
- 31.Jin KL, Pak JH, Park JY, Choi WH, Lee JY, Kim JH, et al. Expression profile of histone deacetylases 1, 2 and 3 in ovarian cancer tissues. J Gynecol Oncol. 2008;19:185–90. doi: 10.3802/jgo.2008.19.3.185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Fang JY, Chen YX, Juan L, Rong L, Li Y, Zhu HY, et al. Epigenetic modification regulates both expression of tumor-associated genes and cell cycle progressing in human colon cancer cell lines: Colo-320 and SW1116. Cell Res. 2004;14:217–26. doi: 10.1038/sj.cr.7290222. [DOI] [PubMed] [Google Scholar]
- 33.Johnson MR, Wang K, Smith JB, Heslin MJ, Diasio RB. Quantitation of dihydropyrimidine dehydrogenase expression by real-time reverse transcription polymerase chain reaction. Analytical Biochem. 2000;278:175–84. doi: 10.1006/abio.1999.4461. [DOI] [PubMed] [Google Scholar]
- 34.Rosato RR, Almenara JA, Dai Y, Grant S. Simultaneous activation of the intrinsic and extrinsic pathways by histone deacetylase (HDAC) inhibitors and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) synergistically induces mitochondrial damage and apoptosis in human leukemia cells. Mol Cancer Ther. 2003;2:1273–84. [PubMed] [Google Scholar]
- 35.Matthews GM, Newbold A, Johnstone RW. Intrinsic and extrinsic apoptotic pathway signaling as determinants of histone deacetylase inhibitor antitumor activity. Adv Cancer Res. 2012;116:165–97. doi: 10.1016/B978-0-12-394387-3.00005-7. [DOI] [PubMed] [Google Scholar]
- 36.Angelucci A, Valentini A, Millimaggi D, Gravina GL, Miano R, Dolo V, et al. Valproic acid induces apoptosis in prostate carcinoma cell lines by activation of multiple death pathways. Anticancer Drugs. 2006;17:1141–50. doi: 10.1097/01.cad.0000236302.89843.fc. [DOI] [PubMed] [Google Scholar]
- 37.Sanaei M, Kavoosi F. Effect of Trichostatin A on Histone Deacetylases 1, 2 and 3, p21Cip1/Waf1/Sdi1, p27Kip1, and p57Kip2 Gene Expression in Breast Cancer SK-BR-3 Cell Line. Asian Pac J Cancer Biol. 2020;5:57–62. doi: 10.31557/APJCP.2020.21.6.1819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Shankar S, Singh TR, Fandy TE, Luetrakul T, Ross DD, Srivastava RK. Interactive effects of histone deacetylase inhibitors and TRAIL on apoptosis in human leukemia cells: involvement of both death receptor and mitochondrial pathways. Int J Mol Med. 2005;16:1125–38. [PubMed] [Google Scholar]
- 39.Inoue S, Riley J, Gant TW, Dyer MJ, Cohen GM. Apoptosis induced by histone deacetylase inhibitors in leukemic cells is mediated by Bim and Noxa. Leukemia. 2007;21:1773–82. doi: 10.1038/sj.leu.2404760. [DOI] [PubMed] [Google Scholar]
- 40.Montani MSG, Granato M, Santoni C, Del Porto P, Merendino N, D’Orazi G, et al. Histone deacetylase inhibitors VPA and TSA induce apoptosis and autophagy in pancreatic cancer cells. Cell Oncol. 2017;40:167–80. doi: 10.1007/s13402-017-0314-z. [DOI] [PubMed] [Google Scholar]
- 41.Mühlethaler-Mottet A, Flahaut M, Bourloud KB, Auderset K, Meier R, Joseph JM, et al. Histone deacetylase inhibitors strongly sensitise neuroblastoma cells to TRAIL-induced apoptosis by a caspases-dependent increase of the pro-to anti-apoptotic proteins ratio. BMC Cancer. 2006;6:1–13. doi: 10.1186/1471-2407-6-214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Meng J, Zhang HH, Zhou CX, Li C, Zhang F, Mei QB. The histone deacetylase inhibitor trichostatin A induces cell cycle arrest and apoptosis in colorectal cancer cells via p53-dependent and-independent pathways. Oncol Rep. 2012;28:384–8. doi: 10.3892/or.2012.1793. [DOI] [PubMed] [Google Scholar]
- 43.Eckschlager T, Plch J, Stiborova M, Hrabeta J. Histone deacetylase inhibitors as anticancer drugs. Int J Mol Sci. 2017;18:1414–21. doi: 10.3390/ijms18071414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Wu LF, Li GP, Su JD, Pu ZJ, Feng JL, Ye YQ, et al. Involvement of NF-κB activation in the apoptosis induced by extracellular adenosine in human hepatocellular carcinoma HepG2 cells. Biochem Cell Biol. 2010;88:705–14. doi: 10.1139/O10-008. [DOI] [PubMed] [Google Scholar]
- 45.Suzuki T, Yokozaki H, Kuniyasu H, Hayashi K, Naka K, Ono S, et al. Effect of trichostatin A on cell growth and expression of cell cycle‐and apoptosis‐related molecules in human gastric and oral carcinoma cell lines. Int J Cancer. 2000;88:992–7. doi: 10.1002/1097-0215(20001215)88:6<992::aid-ijc24>3.0.co;2-9. [DOI] [PubMed] [Google Scholar]
- 46.Decrion-Barthod AZ, Bosset M, Plissonnier ML, Marchini A, Nicolier M, Launay S, et al. Sodium butyrate with UCN-01 has marked antitumour activity against cervical cancer cells. Anticancer Res. 2010;30:4049–61. [PubMed] [Google Scholar]









