(+)‑Terrein inhibits human hepatoma Bel‑7402 proliferation through cell cycle arrest

  • Authors:
    • Fengli Zhang
    • Meiheriguli Mijiti
    • Wei Ding
    • Jiale Song
    • Ying Yin
    • Wei Sun
    • Zhiyong Li
  • View Affiliations

  • Published online on: January 14, 2015     https://doi.org/10.3892/or.2015.3719
  • Pages: 1191-1200
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatoma is a common malignant tumor. Thus, the development of a high‑efficacy therapeutic drug for hepatoma is required. In this study, (+)‑terrein isolated from the marine sponge‑derived Aspergillus terreus PF‑26 against cell growth, apoptosis and cell cycle were assessed by MTT and flow cytometry. mRNA array containing 73 cell cycle‑related genes and three cell morphology‑related genes was generated and its performance evaluated. The cell cycle pathway map was created using the pathview package. The results showed that (+)‑terrein inhibited the growth of Bel‑7402 cells with alterations in cell morphology and a reduced transcript expression of cell morphology genes (fibronectin, N‑cadherin, and vimentin). In addition, flow cytometric analysis revealed that (+)‑terrein arrested the Bel‑7402 cell cycle without inducing apoptosis. Based on multiple mRNA analysis, the downregulated expression of the CCND2, CCNE2, CDKN1C, CDKN2B, ANAPC, PKMYT1, CHEK2 and PCNA genes was observed in 10 µM (+)‑terrein‑treated Bel‑7402 cells (>2‑fold and P≤0.05), compared with the controls. Thus, the antiprolife­rative mechanism of (+)‑terrein against Bel‑7402 cells may be due to the cell cycle arrest by blocking cell cycle gene expression and changing cell morphology.

Introduction

Hepatocellular carcinoma (HCC) is the most common malignant tumor and the third leading cause of cancer mortality worldwide (1). In China, HCC causes ~110,000 deaths annually (2). Current chemotherapeutic drugs for treating HCC are restricted in their clinical application because of toxicity and low efficacy (3,4). Arsenic trioxide (As2O3) (1–8 μM) arrests HCC (HepG2 and SMMC-7721 cells) in the G2/M phase (5). However, applications there of are controversial as arsenic compounds exhibit high toxicity. Therefore, developing a novel high-efficacy therapeutic drug for hepatoma is required (4). In 1935, Raistrick and Smith (6) first isolated (+)-terrein from Aspergillus terreus. Terreins with distinct configurations have received considerable attention because of their various substantial bioactivities, including anticancer properties against human cells (710).

In antitumor therapy, numerous drugs affect tumorigenesis and tumor growth; however, the key is determining which drugs to exploit in the areas of signal transduction, cell-cycle regulation, apoptosis, telomere biology, necrosis, autophagy, cell senescence and angiogenesis (1113). Angiogenesis is a critical process for tumor growth, invasion, and metastasis (14). Arakawa et al (7) determined that (−)-terrein inhibited angiogenin secretion in the tumor angiogenesis of androgen-dependent prostate cancer cells. Apoptosis is a form of cell death (12,15). Furthermore, (+)-terrein suppresses the proliferation of breast cancer cells (9), human cervical carcinoma cells (10), and pulmonary tumor cells (8) by inducing an apoptotic mechanism. These results suggested that terreins inhibit tumor cell growth through multiple strategies.

Findings of studies showed that the isolation and production of (+)-terrein (the molecular formula is C8H10O3 and the molecular weight is 154; Fig. 1) from the fungus A. terreus PF-26 associated with marine sponges were improved to ~9.07 g/l (1619). However, the activity of (+)-terrein against HCC and its mechanism remain unknown. In this study, the anticancer activity and mechanism of (+)-terrein against HCC were investigated using the Bel-7402 human hepatoma cell line. The results showed that (+)-terrein suppressed Bel-7402 human hepatoma cell growth and proliferation. The results indicated that 10 μM (+)-terrein induced cell cycle arrest in the G2/M phase and decreased the cell morphology gene expression of fibronectin, N-cadherin, and vimentin. In addition, the high-throughput platform with parallel detection of multiple mRNAs revealed that treating Bel-7402 cells with (+)-terrein substantially altered the expression of cell cycle-related genes. In addition, (+)-terrein did not induce Bel-7402 cell apoptosis, indicating that (+)-terrein inhibits cell proliferation through distinct mechanisms in different cell strains.

Materials and methods

Reagents and cell lines

(+)-Terrein (Fig. 1) was isolated from A. terreus PF-26, as described previously (18). The isolated (+)-terrein was dissolved in phosphate-buffered saline (PBS, pH 7.2) for subsequent experiments. The human A549 lung adenocarcinoma epithelial cell line was provided by Dr Wei Ma (Shanghai Jiao Tong University, China), and the Bel-7402 human hepatoma cell line was obtained from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China). The mentioned cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) containing 10% fetal bovine serum and 100 U/ml of penicillin and streptomycin. Unless otherwise mentioned, reagents for cell cultures were purchased from Gibco/Invitrogen (New York, Grand Island, USA) and biochemical reagents were obtained from Sigma (New York, NY, USA) or Ameresco (Solon, OH, USA). The cells were grown in a 5% CO2 atmosphere at 37°C.

Cell viability and proliferation

An MTT assay [3-(4,5-dime-thylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] was performed with ~4×103 cells/well in 96-well plates. The plates containing the cells were incubated at 37°C for 24 h, and the cells were treated with (+)-terrein at 37°C for 48 h. The protocol was performed using an MTT cell proliferation and cytotoxicity detection kit (KeyGen, Nanjing, China) according to the manufacturer’s instructions. Briefly, DMEM was supplemented with 50 μl of MTT reagent to each well and incubated at 37°C for 4 h. Thereafter, the MTT solution was removed. Following the addition of 150 μl of dimethyl sulfoxide (DMSO), the plates were incubated at 37°C for 15 min to dissolve the formazan crystals. Absorbance of DMSO extracts was detected at 550 nm by using an Enspire 2300 microplate reader (PerkinElmer, Foster City, CA, USA). A total of ~3×105 cells/well were inoculated in 6-well plates at 37°C for 24 h and treated with (+)-terrein at 37°C for 48 h. The PBS-treated cells served as controls. These cells were used to detect cell morphology, cell cycle, apoptosis, and RNA extraction.

Light microscopy analysis

Cells (3×105) were cultured in 6-well plates at 37°C for 24 h. The Bel-7402 and A549 cells were then treated with 10 μM and 10 mM (+)-terrein at 37°C for 48 h, respectively, and the PBS-treated cells served as controls. The treated cells were used to observe cell morphology and apoptosis. Light microscopy images of the cells were captured using a Nikon Eclipse Ti-inverted microscope and a Nikon digital sight s-Qi1Mc camera (both from Yokohama, Japan). For each surface, three non-overlapping images were selected.

Cell analysis via flow cytometry

The cells were rinsed once in chilled PBS, digested with 0.25% trypsin (Gibco), and then resuspended in DMEM and 10% serum. The suspended cells were centrifuged at 2,000 × g at 4°C for 5 min and washed once in cold PBS. The cells were stained with Alexa Fluor® 488 Annexin V and PI by using an Alexa Fluor® 488 Annexin V/Dead cell apoptosis kit (Invitrogen, New York, USA) according to the manufacturer’s instructions. The stained cells were analyzed using flow cytometry (FACSAria-II, BD Biosciences, San Jose, CA, USA).

Gene expression analysis of cell cycle and cell morphology

The Bel-7402 cells treated with 10 μM (+)-terrein were trypsinized and washed with a PBS buffer. Fifty microliters of single cell suspension was examined using a cell cycle detection kit (KeyGen). The stained cells were analyzed using flow cytometry (FACSAria II).

Total RNA extraction

The Bel-7402 cells treated with (+)-terrein or PBS were trypsinized and washed with a PBS buffer. The cells were collected using centrifugation at 2,000 × g for 5 min. The cell pellet was then resuspended in RL buffer and centrifuged at 13,000 × g for 5 min. Total RNA was extracted according to the manufacturer’s instructions (CWBio, Beijing, China). The purity and concentration of RNA was confirmed by the relative absorbance ratio at 260/280 nm and 260 nm, respectively, by using NanoDrop 2000 (Thermo Fisher Scientific, Waltham, MA, USA).

Reverse transcription

Reverse transcription was performed according to the manufacturer’s instructions (Thermo Fisher Scientific). Total RNA (1 μg) was mixed with 4 μl of a 5X reaction buffer, 1 μl of oligo (dT)18 primer, 1 μl of RiboLock RNase inhibitor, 2 μl of a 10 mM dNTP mix, 1 μl of RevertAid reverse transcriptase (100 U/μl), and then, ddH2O was added to increase the volume to 20 μl. Reverse transcription was performed at 37°C for 60 min and then at 70°C for 5 min. The resulting cDNA was stored at −70°C until use.

Polymerase chain reaction (PCR) array and primer design

Table I shows that 73 potential genes involved in the cell cycle were used as the target mRNAs. GAPDH, b2-MG, β-actin, RPL27, HPRT1, and OAZ1 were used as housekeeping genes for the control cells. The primers were designed (CT Bioscience Co., Changzhou, China) to cover all of the transcripts of each gene. Table I shows the RefSeq accession IDs. All of the primers had a similar melting temperature (Tm), and it was not located in the genomic repetitive regions. The primers were selected based on criteria such as a typical amplification curve and single peak from the post-PCR melting curve. FN, gene ID 2335; N-cadherin, gene ID 1000; and vimentin, gene ID 7431 which are involved in cell morphology were also investigated.

Table I

Gene information detected by the primers.

Table I

Gene information detected by the primers.

Gene ID as in PCR arraySymbolGene IDGene name
1ANAPC229882Anaphase promoting complex subunit 2
2ANAPC429945Anaphase promoting complex subunit 4
3ANAPC551433Anaphase promoting complex subunit 5
4BUB1699Budding uninhibited by benzimidazoles 1 homolog (yeast)
5BUB1B701Budding uninhibited by benzimidazoles 1 homolog β (yeast)
6BUB39184Budding uninhibited by benzimidazoles 3 homolog (yeast)
7CCNE1898Cyclin E1
8CCNE29134Cyclin E2
9CCND1595Cyclin D1
10CCND2894Cyclin D2
11CCND3896Cyclin D3
12CCNH902Cyclin H
13CDC168881Cell division cycle 16 homolog (S. cerevisiae)
14CDC20991Cell division cycle 20 homolog (S. cerevisiae)
15CDC238697Cell division cycle 23 homolog (S. cerevisiae)
16CDC25A993Cell division cycle 25 homolog A (S. pombe)
17CDC25B994Cell division cycle 25 homolog B (S. pombe)
18CDC25C995Cell division cycle 25 homolog C (S. pombe)
19CDC26246184Cell division cycle 26 homolog (S. cerevisiae)
20CDC27996Cell division cycle 27 homolog (S. cerevisiae)
21CDC6990Cell division cycle 6 homolog (S. cerevisiae)
22CDC78317Cell division cycle 7 homolog (S. cerevisiae)
23CDK41019Cyclin-dependent kinase 4
24CDK61021Cyclin-dependent kinase 6
25CDK71022Cyclin-dependent kinase 7
26CDKN1B1027Cyclin-dependent kinase inhibitor 1B (p27, Kip1)
27CHEK11111Checkpoint kinase 1
28CHEK211200Checkpoint kinase 2
29E2F11869E2F transcription factor 1
30E2F21870E2F transcription factor 2
31E2F31871E2F transcription factor 3
32HDAC13065Histone deacetylase 1
33MAD2L14085MAD2 mitotic arrest deficient-like 1 (yeast)
34MAX4149MYC-associated factor X
35MCM24171Minichromosome maintenance complex component 2
36MCM34172Minichromosome maintenance complex component 3
37MCM44173Minichromosome maintenance complex component 4
38MCM54174Minichromosome maintenance complex component 5
39MCM64175Minichromosome maintenance complex component 6
40MCM74176Minichromosome maintenance complex component 7
41ORC1L4998Origin recognition complex, subunit 1
42ORC2L4999Origin recognition complex, subunit 2
43ORC6L23594Origin recognition complex, subunit 6
44PCNA5111Proliferating cell nuclear antigen
45PKMYT19088Protein kinase, membrane-associated tyrosine/threonine 1
46RB15925Retinoblastoma 1
47RBL15933Retinoblastoma-like 1 (p107)
48SKP26502S-phase kinase-associated protein 2, E3 ubiquitin protein ligase
49SMC1A8243Structural maintenance of chromosomes 1A
50TOP2A7153Topoisomerase (DNA) II α 170 kDa
51TP537157Tumor protein p53
52TFDP17027Transcription factor Dp-1
53WEE17465WEE1 homolog (S. pombe)
54Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation
YWHAE7531Protein, ɛ polypeptide
55CDKN2A1029Cyclin-dependent kinase inhibitor 2A
56CDKN2B1030Cyclin-dependent kinase inhibitor 2B (p15, inhibits CDK4)
57PSMD95715Proteasome (prosome, macropain) 26S subunit, non-ATPase, 9
58CDKN1C1028Cyclin-dependent kinase inhibitor 1C (p57, Kip2)
59CDC2983Cyclin-dependent kinase 1
60CCNB29133Cyclin B2
61CCNB1891Cyclin B1
62CDKN1A1026Cyclin-dependent kinase inhibitor 1A (p21, Cip1)
63CDK21017Cyclin-dependent kinase 2
64CCNA18900Cyclin A1
65CCNA2890Cyclin A2
66MYC4609V-myc myelocytomatosis viral oncogene homolog (avian)
67CDKN2C1031Cyclin-dependent kinase inhibitor 2C (p18, inhibits CDK4)
68GAPDH2597 Glyceraldehyde-3-phosphate dehydrogenase
69ACTB60Actin, β
70B2M567 β-2-microglobulin
71HPRT13251Hypoxanthine phosphoribosyltransferase 1
72OAZ14946Ornithine decarboxylase antizyme 1
73RPL276155Ribosomal protein L27
Real-time PCR analysis

Each cDNA was diluted to 1 ml with ddH2O and mixed with 1 ml of 2X SYBR Premix Ex Taq™ (Takara, Dalian, China). Twenty microliters of this mixture was added to each well of a 96-well PCR array, except for genomic DNA control (GDC). The 96-well PCR plates containing gene primers were prepared by the CT Bioscience Company (Changzhou, China). The sealed PCR plate was loaded in an Eppendorf Realplex 4S (Hamburg, German). The PCR was performed under the following conditions: 95°C for 5 min, 40 cycles at 95°C for 15 sec, 60°C for 15 sec, and 72°C for 20 sec. The melting curve procedure (95°C for 15 sec, 60°C for 15 sec, and 95°C for 15 sec) was implemented to analyze the PCR specificity. Dissociation curves (DC) and melting temperatures (Tm) were recorded. Relative changes in gene expression were calculated using the threshold cycle (Ct) method (20). The formula used is presented as follows: n-fold change = 2−ΔΔCt = (Ct target gene − Ct internal control gene) treated sample − (Ct target gene − Ct internal control gene) control sample.

Statistical and pathway analysis

Statistical product and service solutions (SPSS ver. 13.0) was used for the data analysis. All of the experiments were conducted in duplicate. The results are presented as mean ± SD (standard deviation) unless otherwise specified. The P-values were two-tailed. P≤0.05 was considered to indicate a statistically significant difference. The cell-cycle pathway is a highly regulated process that incorporates three major checkpoints including the participation of several genes (21). The functional pathways associated with the set of differentially expressed genes were analyzed using the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis (http://www.kegg.jp/kegg/pathway.html). Differentially expressed gene with >2-fold change were analyzed in the cell-cycle pathway (22). The pathway map was created using the Pathview™ package (23).

Results

(+)-Terrein reduces cell growth

The in vitro toxicity of (+)-terrein against Bel-7402 cells and the A549 human lung adenocarcinoma epithelial cell line was evaluated using the MTT method [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide] to determine the potential inhibitory concentration. MTT analysis revealed that(+)-terrein inhibited cell viability and proliferation in a concentration-dependent manner. Fig. 2A shows the inhibition of Bel-7402 cells at various (+)-terrein concentrations. The IC50 (half maximal inhibitory concentration) value of Bel-7402 was calculated as 11.63 μM ±0.02. The dose-dependent inhibition of Bel-7402 indicated that 1 μM (+)-terrein was non-toxic and 10 μM (+)-terrein was cytotoxic and associated with a survival rate of 57%. The survival rate of the Bel-7402 cells treated with 100 μM and 1 mM (+)-terrein exhibited inhibitory activities of ~22 and 13%, respectively. A low dose of (+)-terrein did not substantially affect the A549 cells. As indicated in Fig. 2B, (+)-terrein at various concentrations induced inhibitory activity against the A549 cells. (+)-Terrein at concentrations of 10 μM, 100 μM, and 1 mM exhibited weak inhibitory activity, and the survival rate of the treated cells was 91, 82, and 72%, respectively. Moreover, (+)-terrein at 10 mM was cytotoxic to A549 cells, and the survival rate of the treated cells was 52%.

(+)-Terrein induces cell morphology change

We observed a marked phenomenon regarding cell morphology alterations when cells were treated with (+)-terrein. Morphological changes in the Bel-7402 cells exposed to 10 μM (+)-terrein and in the A549 cells exposed to 10 mM (+)-terrein for 48 h were examined using light microscopy (Fig. 3). The Bel-7402 cell morphology alterations were from epithelial-like to spherical, when the cells were treated with 10 μM (+)-terrein (Fig. 3A and B). The morphological changes from epithelial-like to spherical of the A549 cells exposed to 10 mM (+)-terrein for 48 h were examined using light microscopy (Fig. 3C and D). However, 10 μl of (+)-terrein did not significantly affect the morphology of A549 cells (data not shown). The cells were adherent, and all of the experiments were repeated at least five times.

The cell morphology gene (FN, N-cadherin, and vimentin) expression of the Bel-7402 cells treated with 10 μM (+)-terrein was investigated. The results showed that the gene expression of FN, N-cadherin, and vimentin decreased −3.61-, −2.39-, and −2.05-fold, respectively, compared with the control (Table II).

Table II

Downregulated expression of cell morphology genes in the Bel-7402 cells treated with (+)-terrein.

Table II

Downregulated expression of cell morphology genes in the Bel-7402 cells treated with (+)-terrein.

Gene nameGene IDPrimersExpression fold
FN (fibronectin)2335F, FN1: 5′-AACCTCGGCTTCCTCCATAA-3′
R, FN1: 5′-AACAGTGGGAGCGGACCTA-3′
−3.61
VIM (vimentin)7431F, VIM: 5′-GCCAACCGGAACAATGAC-3′
R, VIM: 5′-GTGAGGGACTGCACCTGTCT-3′
−2.05
CDH2 (N-cadherin)1000F, CDH2: 5′-CTAACCCGTCGTTGCTGTTT-3′
R, CDH2: 5′-ACAGAATCAGTGGCGGAGAT-3′
−2.39

[i] The Bel-7402 cells were treated with 10 μM (+)-terrein for 48 h. Each value is the mean of three independent experiments. P≤0.05 and were statistically significant. F, forward; R, reverse.

(+)-Terrein inhibits cell apoptosis and necrosis

To determine whether this reduction in cell growth induced by (+)-terrein was mediated by apoptosis, flow cytometric analysis was performed using PI and Annexin-V staining. The results indicated that the apoptotic levels of Bel-7402 and A549 cells treated with (+)-terrein (10 μM and 10 mM) did not increase (Fig. 4), but the number of cells treated with (+)-terrein was less than that of the control cells, according to cell counting (data not shown). The mean percentage ± SD (n=3) of early apoptosis for the Bel-7402 and A549 control cells was 5.33±1.05 and 6.17±0.93, respectively, but that for the Bel-7402 and A549 cells after treatment with (+)-terrein for 48 h was 2.26±0.50 and 0.53±0.31, respectively (Table III). Fig. 4 shows that (+)-terrein at 10 μM and 10 mM inhibited early apoptosis. As indicated in Table III, the mean percentage ± SD (n=3) of late apoptosis and necrosis for the Bel-7402 and A549 control cells was 1.77±0.61 and 5.43±2.55, respectively, but that for the Bel-7402 and A549 cells following treatment with (+)-terrein for 48 h was 0.83±0.23 and 1.93±1.07, respectively (Table III). Based on these results, (+)-terrein inhibited late cell apoptosis and necrosis (Fig. 4). Standard deviation was larger than the average deviation; however, for every independent experiment, the percentage value (%) of apoptosis and necrosis in the treated Bel-7402 and A549 cells was lower than that in the control cells.

Table III

(+)-Terrein induced cell apoptosis and necrosis.

Table III

(+)-Terrein induced cell apoptosis and necrosis.

Apoptosis (%)Necrosis (%)


CellsControlTreated cellsControlTreated cells
Bel-74025.33±1.052.26±0.501.77±0.610.83±0.23
A45496.17±0.930.53±0.315.43±2.551.93±1.07

[i] Early apoptotic cells (%) were decreased after treatment with (+)-terrein at 10 μM to Bel-7402 or at 10 mM to A549 for 48 h. Cells not treated with (+)-terrein served as controls. Late apoptotic and necrotic cells (%) were decreased after treatment with (+)-terrein at 10 μM to Bel-7402 or at 10 mM to A549 for 48 h. Each value is the mean ± SD of five independent experiments.

Effect of (+)-terrein on Bel-7402 cell cycle

Cell proliferation depends on the specific progression of the cell cycle (21). Thus, the cell cycle was analyzed to investigate the anti-proliferative mechanism of (+)-terrein against Bel-7402 cells. Provided the influence of (+)-terrein on A549 occurs at an exceedingly high concentration (>mmol), the anti-proliferative mechanism of (+)-terrein against A549 was not examined. When the Bel-7402 cells were treated with 10 μM (+)-terrein for 48 h, the proportion of the cells in the G0/G1 and S phases was reduced, whereas the proportion of cells in the G2/M phase was increased (Table IV). This result suggested that the cell cycle was arrested by (+)-terrein. Thus, (+)-terrein might decrease Bel-7402 cell growth by inducing cell cycle arrest.

Table IV

Effect of (+)-terrein on the cell cycle of the Bel-7402 cells.

Table IV

Effect of (+)-terrein on the cell cycle of the Bel-7402 cells.

Sample G0/G1 (%)S (%)G2/M (%)
Control59.91±2.835.29±2.14.8±0.53
Treated cells (10 μM)56.88±3.535.26±2.87.36±0.27

[i] The Bel-7402 cells were treated with (+)-terrein at 10 μM for 48 h. Cells not treated with (+)-terrein served as controls. Each value is the mean ± SD of three independent experiments.

Effect of (+)-terrein on cell-cycle regulators

In this study, high-throughput gene expression analysis of 73 genes was performed (Table I). Cell cycle-related gene expression was performed by comparing the gene expression between cDNA samples of (+)-terrein-treated Bel-7402 cells and control cells. Melting curve analysis was used to assess the specificity of the array. A single product peak observed from each reaction without secondary products indicated a high specificity of PCR assay (data not shown). A subset of differentially expressed genes involved in the cell cycle was selected from all the microarray data by performing initial filtration of the P-value (P≤0.05) and expression level (>2-fold) of the 10 μM (+)-terrein-treated cells. The cell-cycle scheme from the KEGG database (http://www.kegg.jp/kegg/pathway.html) was presented. Downregulated genes were labeled in green, while no upregulated genes were overexpressed (Fig. 5). Compared with the control group, the average expression values of CCND2, CCNE2, CDKN1C, CDKN2B, ANAPC5, PKMYT1, CHEK2, and PCNA genes in the 10 μM (+)-terrein-treated group were significantly decreased by 5.52-, 3.30-, 5.32-, 2.24-, 2.52-, 2.52-, 2.31-, and 2.10-fold (>2-fold; P≤0.05), respectively (Table V), and the expression level of the remaining 65 genes was evidently unchanged (data not shown). Eight obviously downregulated genes were visualized in the cell-cycle pathway (Fig. 5). Moreover, the results of flow cytometry (Table IV) indicated that (+)-terrein arrested the cell cycle.

Table V

Downregulated cell-cycle genes in the Bel-7402 cells treated with (+)-terrein.

Table V

Downregulated cell-cycle genes in the Bel-7402 cells treated with (+)-terrein.

Gene no.Gene nameFunction in cell cyclen-fold
1CCND2G1 phase and G1/S transition−5.52
2CCNE2G1/S checkpoint−3.30
3CDKN1CCauses arrest of the cell cycle in G1 phase−5.32
4CDKN2BCell-cycle checkpoint and cell-cycle arrest−2.24
5ANAPC5G2 phase and G2/M transition−2.52
6PKMYT1Protein kinase, membrane-associated tyrosine/threonine 1−2.52
7CHEK2Checkpoint kinase 2−2.31
8PCNAProliferating cell nuclear antigen−2.10

[i] Bel-7402 cells treated with 10 μM (+)-terrein for 48 h and cells not treated with (+)-terrein served as controls. Each value is the mean of three independent experiments.

Discussion

Numerous drugs affect tumorigenesis and tumor growth through several mechanisms, including signal transduction, cell-cycle regulation, apoptosis, telomere biology, angiogenesis and cell senescence (11,13). Most available cancer drugs are antimitotic and act by interfering with the basic mechanism of DNA synthesis and cell division (20). (+)-Terrein inhibited cell growth at various concentrations in various human tumor cell strains (810). The IC50 value of (+)-terrein against human breast cancer MCF-7 cells was 1.1 nM (9). The IC50 value was 0.29 mM for HeLa cells (10) and 0.3 mM for NCI-H292 (8). Strese et al (24) observed that the chemosensitivity of various cell lines was differentially expressed, indicating that distinct cell types with distinct genetic backgrounds exhibit distinct responses to drug treatment (15,25).

Since the adhesion and distribution of anchorage-dependent cells are prerequisites for cell viability and proliferation, cell growth and survival also depend on cell morphology (26). Changes in the synthesis and structure of actin induce changes in cell morphology. FN is a ubiquitous extracellular matrix glycoprotein assembled into an FN matrix in all tissues and throughout all stages of life. Loss of an FN matrix causes changes in cell morphology, cell signaling, proliferation, and cell-cycle progression (27,28). The aberrant expression of N-cadherin by cancer cells contributes to invasiveness and metastasis by making the cells more motile (29). Vimentin is responsible for maintaining cell shape, adhesion and motility (30).

The inhibitory mechanism of (+)-terrein against the Bel-7402 cell differed from the apoptosis of breast cancer and pulmonary tumor cell lines (9,10). Liao et al (9) and Porameesanaporn et al (10) reported that (+)-terrein suppressed the growth of breast cancer and HeLa cancer cell lines by inducing apoptosis. Demasi et al (8) determined that various ranges of (+)-terrein induced pulmonary tumor cell apoptosis through protease inhibitors.

Kim et al (31) determined that (±)-terrein inhibited human epidermal keratinocyte proliferation through extracellular signal-regulated protein kinase inactivation and G2/M cell-cycle arrest. The fundamental task of the cell cycle is to ensure that DNA is successfully replicated in the S phase and that the identical chromosomal copies are equally distributed between two daughter cells in the M phase (32,33). Cell proliferation depends on the progression of the cell cycle through the G0/G1 phase to the S phase (34). In some biological systems, cell-cycle delay and long-term arrest in the G2 phase are well documented, but most variation in cell-cycle duration among tissues is due to variability in the length of the G1 phase (32,33). We determined that (+)-terrein decreased the proportion of the Bel-7402 cells in the G0/G1 and S phases (Table IV). The data suggested that (+)-terrein delayed the progression of the cell cycle. The results indicated that 10 μM (+)-terrein inhibited Bel-7402 cell growth (Fig. 2) by arresting the cell cycle in the G2/M phase (Table IV). The sensitivity of the drug-induced DNA damage was, not only associated with the interaction between drug and target, but also depended on the dose, time (15) and cellular status (34). The results of this study confirmed that various strains (Bel-7402 and A549) exhibit a distinct response to (+)-terrein (Fig. 2). Therefore, the anti-proliferative mechanism of (+)-terrein based on distinct drug doses, time and cellular status remains to be investigated.

Cell-cycle progression in mammalian cells is regulated by various proteins (35). Cell proliferation is closely controlled by positive and negative regulators that determine cell progress throughout the cell cycle (36). The cyclin/cyclin-dependent kinase (Cdk) complexes and Cdk inhibitors (CDKIs) are crucial regulators of cell-cycle progression (37).

Eight genes are related to the cell cycle (Fig. 5). The CCND2 (38), CCNE2 (39), CDKN1C (40) and CDKN2B (41) genes, positively regulated the G1 and G1/S phases of the cell cycle, whereas the APC gene negatively regulated the G2/M transition in the cell cycle (42). Distinct cyclins exhibited distinct expression and degradation patterns, which contribute to the temporal coordination of each mitotic event (38). Cyclin D encoded by the CCND2 gene forms a complex regulatory subunit of CDK4 or CDK6, the activity of which is required for cell-cycle G1/S transition (38). Cyclin E encoded by the CCNE2 gene controls the G1- to S-phase transition in the cell cycle (39). Cyclin-dependent kinase inhibitor 1C (p57, Kip2), also known as CDKN1C, causes cell cycle arrest in the G1 phase (40). Cyclin-dependent kinase 4 inhibitor B (ink4b) encoded by the CDKN28B gene is a potential factor of cell cycle arrest in the G1 phase (41). In addition, anaphase-promoting complex subunit 5 (ANAPC5) consists of at least eight protein subunits, including APC5, CDC27 (APC3; MIM 116946), CDC16 (APC6; MIM 603461), and CDC23 (APC8; MIM 603462). The APC/C targets the mitotic cyclins for degradation, resulting in the inactivation of mitotic cyclin-dependent kinase (M-CdK) complexes, promoting exit from mitosis and cytokinesis (42). Although distinct processes are responsible for this inhibition, a crucial process is the activation of the APC/C by Cdh1. This continued activation prevents the accumulation of cyclin, which triggers another round of mitosis instead of exiting (42). The results from the multiple mRNA analysis further proved that the Bel-7402 cells were arrested in the G2/M phase by 10 μM (+)-terrein.

Membrane-associated tyrosine- and threonine-specific CDC2-inhibitory kinase encoded by the PKMYT1 gene negatively regulates cell-cycle G2/M transition (43). A decrease in the expression of PKMYT1 (Table V) and ANAPC (Fig. 5 and Table V) genes was conducive to arresting the cells in the G2/M phase of the cell cycle. The CHEK2 gene provides instructions for producing checkpoint kinase 2 (CHK2). CHEK2 activation in response to DNA damage prevents the cell from entering mitosis (44). Proliferating cell nuclear antigen (PCNA) is a nuclear protein that acts as a processivity factor for DNA polymerase ɛ in eukaryotic cells (45). The expression levels of PCNA varied throughout the cell-cycle progression, and the maximal expression was observed in the G1 and S phases (46). Furthermore, the PCNA expression level was downregulated as the cells exited the cell cycle and differentiated (47). Based on the PCNA expression level characteristics (46,47), we obtained consistent results indicating that 10 μM (+)-terrein increased the proportion of treated cells in the G2/M phase (Table IV) and decreased the PCNA expression 2.10-fold, compared with the control group (Table V). Thus, (+)-terrein induced cell cycle arrest by breaking down the balance between multiple gene expressions of the cell cycle.

In conclusion, (+)-terrein exhibited cytotoxicity against the Bel-7402 human hepatoma cell line, yielding an IC50 value of 11.63 μM ±0.02. In addition, (+)-terrein induced round-cell morphology of Bel-7402 and A549 cells but did not induce cell apoptosis. Furthermore, (+)-terrein inhibited Bel-7402 human hepatoma cell proliferation and arrested the cell cycle in the G2/M phase by breaking down the expression of the CCND2, CCNE2, CDKN1C, CDKN2B, ANAPC5, PKMYT1, CHEK2 and PCNA cell cycle-related genes. The results suggest that (+)-terrein inhibited human tumor cell growth through various strategies. The potential application of (+)-terrein remains to be investigated in future studies.

Acknowledgements

We would like to thank Dr Qian Luo at the Instrument Sharing and Technical Service Platform of SJTU for technical advice on using flow cytometry. We would also like to thank Dr Valliappan Karuppiah for providing assistance with the language. This study was supported by the High-Tech Research and Development Program of China (2011AA090702), the Medical and Engineering Cross Funds of Shanghai Jiao Tong University (YG2011ms13), and the National Natural Science Foundation of China (J1210047 and 31300104).

Abbreviations:

ANAPC5

anaphase-promoting complex subunit 5

Cdk

cyclin/cyclin-dependent kinase

CDKI

Cdk inhibitors

CDKN1C

cyclin-dependent kinase inhibitor 1C

Ct

threshold cycle

DC

dissociation curves

GDC

genomic DNA control

HCC

hepatocellular carcinoma

ink4b

cyclin-dependent kinase 4 ihibitor B

M-CdK

mitotic cyclin-dependent kinase

PCNA

proliferating cell nuclear antigen

SPSS

Statistical Product and Service Solutions

Tm

melting temperature

KEGG

Kyoto Encyclopedia of Genes and Genomes

References

1 

Schütte K, Bornschein J and Malfertheiner P: Hepatocellular carcinoma - epidemiological trends and risk factors. Dig Dis. 27:80–92. 2009. View Article : Google Scholar

2 

Xie SL, Zhu MG, Lv GY, Zhang Q and Wang GY: The role of RhoC in the proliferation and apoptosis of hepatocellular carcinoma cells. Med Oncol. 29:1802–1809. 2012. View Article : Google Scholar

3 

Cao H, Phan H and Yang LX: Improved chemotherapy for hepatocellular carcinoma. Anticancer Res. 32:1379–1386. 2012.PubMed/NCBI

4 

Nouso K: Current chemotherapies for advanced hepatocellular carcinoma. Clin J Gastroenterol. 6:89–93. 2013. View Article : Google Scholar

5 

Zhang X, Jia S, Yang S and Yang Y, Yang T and Yang Y: Arsenic trioxide induces G2/M arrest in hepatocellular carcinoma cells by increasing the tumor suppressor PTEN expression. J Cell Biochem. 113:3528–3535. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Raistrick H and Smith G: Studies in the biochemistry of micro-organisms: The metabolic products of Aspergillus terreus Thom. A new mould metabolic product-terrein. Biochem J. 29:606–611. 1935.PubMed/NCBI

7 

Arakawa M, Someno T, Kawada M and Ikeda D: A new terrein glucoside, a novel inhibitor of angiogenin secretion in tumor angiogenesis. J Antibiot. 61:442–448. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Demasi M, Felicio AL, Pacheco AO, Leite HG, Lima C and Andrade LH: Studies on terrein as a new class of proteasome inhibitors. J Braz Chem Soc. 21:299–305. 2010. View Article : Google Scholar

9 

Liao WY, Shen CN, Lin LH, Yang YL, Han HY, Chen JW, Kuo SC, Wu SH and Liaw CC: Asperjinone, a nor-neolignan, and terrein, a suppressor of ABCG2-expressing breast cancer cells, from thermophilic Aspergillus terreus. J Nat Prod. 75:630–635. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Porameesanaporn Y, Uthaisang-Tanechpongtamb W, Jarintanan F, Jongrungruangchok S and Thanomsub Wongsatayanon B: Terrein induces apoptosis in HeLa human cervical carcinoma cells through p53 and ERK regulation. Oncol Rep. 29:1600–1608. 2013.PubMed/NCBI

11 

Anisimov VN: Biology of aging and cancer. Cancer Control. 14:23–31. 2007.PubMed/NCBI

12 

Edinger AL and Thompson CB: Death by design: apoptosis, necrosis and autophagy. Curr Opin Cell Biol. 16:663–669. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Gibbs JB: Mechanism-based target identification and drug discovery in cancer research. Science. 287:1969–1973. 2000. View Article : Google Scholar : PubMed/NCBI

14 

Bergers G and Benjamin LE: Tumorigenesis and the angiogenic switch. Nat Rev Cancer. 3:401–410. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Schmitt CA and Lowe SW: Apoptosis and therapy. J Pathol. 187:127–137. 1999. View Article : Google Scholar : PubMed/NCBI

16 

Xiao L, Yin Y, Sun W, Zhang F, Zhang F and Li Z: Enhanced production of (+)-terrein by Aspergillus terreus strain PF26 with epigenetic modifier suberoylanilide hydroxamic acid. Proc Biochem. 48:1635–1639. 2013. View Article : Google Scholar

17 

Xu B, Yin Y, Zhang F, Li Z and Wang L: Operating conditions optimization for (+)-terrein production in a stirred bioreactor by Aspergillus terreus strain PF-26 from marine sponge Phakellia fusca. Bioprocess Biosyst Eng. 35:1651–1655. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Yin Y, Gao Q, Zhang F and Li Z: Medium optimization for the high yield production of single (+)-terrein by Aspergillus terreus strain PF-26 derived from marine sponge Phakellia fusca. Process Biochem. 47:887–891. 2012. View Article : Google Scholar

19 

Yin Y, Xu B, Li Z and Zhang B: Enhanced production of (+)-terrein in fed-batch cultivation of Aspergillus terreus strain PF-26 with sodium citrate. World J Microbiol Biotechnol. 29:441–446. 2013. View Article : Google Scholar

20 

Schmittgen TD and Livak KJ: Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 3:1101–1108. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Evan GI and Vousden KH: Proliferation, cell cycle and apoptosis in cancer. Nature. 411:342–348. 2001. View Article : Google Scholar : PubMed/NCBI

22 

Kanehisa M and Goto S: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28:27–30. 2000. View Article : Google Scholar

23 

Luo W and Brouwer C: Pathview: an R/Bioconductor package for pathway-based data integration and visualization. Bioinformatics. 29:1830–1831. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Strese S, Fryknäs M, Larsson R and Gullbo J: Effects of hypoxia on human cancer cell line chemosensitivity. BMC Cancer. 13:3312013. View Article : Google Scholar : PubMed/NCBI

25 

Evan G and Littlewood T: A matter of life and cell death. Science. 281:1317–1322. 1998. View Article : Google Scholar : PubMed/NCBI

26 

French PW, Donnellan M and McKenzie DR: Electromagnetic radiation at 835 MHz changes the morphology and inhibits proliferation of a human astrocytoma cell line. Bioelectrochem Bioenerg. 43:13–l8. 1997. View Article : Google Scholar

27 

Bourdoulous S, Orend G, MacKenna DA, Pasqualini R and Ruoslahti E: Fibronectin matrix regulates activation of RHO and CDC42 GTPases and cell cycle progression. J Cell Biol. 143:267–276. 1998. View Article : Google Scholar : PubMed/NCBI

28 

Yi M and Ruoslahti E: A fibronectin fragment inhibits tumor growth, angiogenesis, and metastasis. Proc Natl Acad Sci USA. 98:620–624. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Ramis-Conde I, Chaplain MAJ, Anderson ARA and Drasdo D: Multi-scale modelling of cancer cell intravasation: the role of cadherins in metastasis. Phys Biol. 6:0160082009. View Article : Google Scholar : PubMed/NCBI

30 

Mendez MG, Kojima S and Goldman RD: Vimentin induces changes in cell shape, motility, and adhesion during the epithelial to mesenchymal transition. FASEB J. 24:1838–1851. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Kim DS, Lee HK, Park SH, Lee S, Ryoo IJ, Kim WG, Yoo ID, Na JI, Kwon SB and Park KC: Terrein inhibits keratinocyte proliferation via ERK inactivation and G2/M cell cycle arrest. Exp Dermatol. 17:312–317. 2008. View Article : Google Scholar

32 

Heichman KA and Roberts JM: Rules to replicate by. Cell. 79:557–562. 1994. View Article : Google Scholar : PubMed/NCBI

33 

Nilsson I and Hoffmann I: Cell cycle regulation by the Cdc25 phosphatase family. Prog Cell Cycle Res. 4:107–114. 2000. View Article : Google Scholar : PubMed/NCBI

34 

Gonzalez VM, Fuertes MA, Alonso C and Perez JM: Is cisplatin-induced cell death always produced by apoptosis? Mol Pharmacol. 59:657–663. 2001.PubMed/NCBI

35 

Sherr CJ: Cancer cell cycles. Science. 274:1672–1677. 1996. View Article : Google Scholar : PubMed/NCBI

36 

Brooks G and La Thangue NB: The cell cycle and drug discovery: the promise and the hope. Drug Discov Today. 4:455–464. 1999. View Article : Google Scholar : PubMed/NCBI

37 

Nurse P: A long twentieth century of the cell cycle and beyond. Cell. 100:71–78. 2000. View Article : Google Scholar : PubMed/NCBI

38 

Mullany LK, White P, Hanse EA, Nelsen CJ, Goggin MM, Mullany JE, Anttila CK, Greenbaum LE, Kaestner KH and Albrecht JH: Distinct proliferative and transcriptional effects of the D-type cyclins in vivo. Cell Cycle. 7:2215–2224. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Lauper N, Beck AR, Cariou S, Richman L, Hofmann K, Reith W, Slingerland JM and Amati B: Cyclin E2: a novel CDK2 partner in the late G1 and S phases of the mammalian cell cycle. Oncogene. 17:2637–2643. 1998. View Article : Google Scholar : PubMed/NCBI

40 

Matsuoka S, Edwards MC, Bai C, Parker S, Zhang P, Baldini A, Harper JW and Elledge SJ: p57KIP2, a structurally distinct member of the p21CIP1 Cdk inhibitor family, is a candidate tumor suppressor gene. Genes Dev. 9:650–662. 1995. View Article : Google Scholar : PubMed/NCBI

41 

Hannon GJ and Beach D: pl5INK4B is a potential effector of TGF-beta-induced cell cycle arrest. Nature. 371:257–261. 1994. View Article : Google Scholar : PubMed/NCBI

42 

Kraft C, Herzog F, Gieffers C, Mechtler K, Hagting A, Pines J and Peters JM: Mitotic regulation of the human anaphase-promoting complex by phosphorylation. EMBO J. 22:6598–6609. 2003. View Article : Google Scholar : PubMed/NCBI

43 

Liu F, Rothblum-Oviatt C, Ryan CE and Piwnica-Worms H: Overproduction of human Myt1 kinase induces a G2 cell cycle delay by interfering with the intracellular trafficking of Cdc2-cyclin B1 complexes. Mol Cell Biol. 19:5113–5123. 1999.PubMed/NCBI

44 

Cybulski C, Górski B, Huzarski T, Masojć B, Mierzejewski M, Debniak T, Teodorczyk U, Byrski T, Gronwald J, Matyjasik J, Złowocka E, Lenner M, Nej K, Castaneda J, Medrek K, Szymańska A, Szymańska J, Kurzawski G, Suchy J, Oszurek O, Witek A, Narod SA and Lubinski J: CHEK2 is a multiorgan cancer susceptibility gene. Am J Hum Genet. 75:1131–1135. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Kisielewska J, Lu P and Whitaker M: GFP-PCNA as an S-phase marker in embryos during the first and subsequent cell cycles. Biol Cell. 97:221–229. 2005. View Article : Google Scholar

46 

Kumar D, Minocha N, Rajanala K and Saha S: The distribution pattern of proliferating cell nuclear antigen in the nuclei of Leishmania donovani. Microbiology. 155:3748–3757. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Barton KM and Levine EM: Expression patterns and cell cycle profiles of PCNA, MCM6, cyclin D1, cyclin A2, cyclin B1, and phosphorylated histone H3 in the developing mouse retina. Dev Dynam. 237:672–682. 2008. View Article : Google Scholar

Related Articles

Journal Cover

March-2015
Volume 33 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang F, Mijiti M, Ding W, Song J, Yin Y, Sun W and Li Z: (+)‑Terrein inhibits human hepatoma Bel‑7402 proliferation through cell cycle arrest. Oncol Rep 33: 1191-1200, 2015
APA
Zhang, F., Mijiti, M., Ding, W., Song, J., Yin, Y., Sun, W., & Li, Z. (2015). (+)‑Terrein inhibits human hepatoma Bel‑7402 proliferation through cell cycle arrest. Oncology Reports, 33, 1191-1200. https://doi.org/10.3892/or.2015.3719
MLA
Zhang, F., Mijiti, M., Ding, W., Song, J., Yin, Y., Sun, W., Li, Z."(+)‑Terrein inhibits human hepatoma Bel‑7402 proliferation through cell cycle arrest". Oncology Reports 33.3 (2015): 1191-1200.
Chicago
Zhang, F., Mijiti, M., Ding, W., Song, J., Yin, Y., Sun, W., Li, Z."(+)‑Terrein inhibits human hepatoma Bel‑7402 proliferation through cell cycle arrest". Oncology Reports 33, no. 3 (2015): 1191-1200. https://doi.org/10.3892/or.2015.3719