Potential antitumor agent from the endophytic fungus Pestalotiopsis photiniae induces apoptosis via the mitochondrial pathway in HeLa cells

  • Authors:
    • Chuan Chen
    • Shu-Yuan Hu
    • Du-Qiang Luo
    • Si-Yu Zhu
    • Chuan-Qi Zhou
  • View Affiliations

  • Published online on: July 17, 2013     https://doi.org/10.3892/or.2013.2618
  • Pages: 1773-1781
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

4-(3',3'-Dimethylallyloxy)-5-methyl-6-methoxy­phthalide (DMMP) has previously been isolated from the endophytic fungus Pestalotiopsis photiniae. Although the cytotoxic activities of DMMP have been reported, little is known concerning the molecular mechanism of its cytotoxic effect. In the present study, we investigated the effect of DMMP on the growth of several types of cancer cell lines and investigated the mechanism of its antiproliferative effect. DMMP caused the growth inhibition of human cancer lines HeLa, MCF7 and MDA-MB-231, but had little antiproliferative effect on MRC5 normal lung cells. DMMP also significantly caused cell cycle arrest in the G1 phase and upregulated the cyclin-dependent kinase inhibitor p27KIPI protein in the HeLa cells. Moreover DMMP was able to induce marked nuclear apoptotic morphology in HeLa cells. DMMP induced apoptosis and loss of mitochondrial membrane potential (ΔΨm) in the HeLa cells. Although the activated forms of caspase-9 and -3 in HeLa cells were detected, pretreatment with caspase inhibitors (Ac-DEVD-CHO and Z-VAD-FMK) failed to attenuate DMMP-induced cell death. In addition, protein levels of the p53 family members, p53 and p73, were upregulated, and DMMP significantly increased the mRNA expression of pro-apoptotic Bcl-2 family genes (PUMA, NOXA, Bax, Bad and Bim). HPV E6-E7 mRNA levels were reduced. In conclusion, DMMP demonstrates potential for use in the treatment of cervical cancer.

Introduction

Natural products and their derivatives have historically been invaluable as sources of chemopreventive and therapeutic agents (1). Recently, more attention has been paid to bioactive natural products of endophytic fungi, isolated from higher plants (2). Endophytic fungi are important components of microbial biodiversity. Biological diversity means chemical diversity implying unique structural features and pronounced biological activities as lead structures for novel drugs (3).

Apoptotic cell death is a self-destructing process under strict control and plays an important role in tissue homeostasis. Impairment of the apoptotic signaling pathway leads to insufficient or excessive cell death which results in the progression of human cancers (4). Thus, a chemical compound which promotes cell apoptosis and inhibits cancer cell proliferation is considered to be a potential anticancer drug (5).

The p53 family members are mainly involved in apoptosis induction and cell cycle arrest under cellular stress. The p53 family consists of 3 members: p53, p63 and p73. Tumor suppressor p53 is a transcription factor that maintains genome stability and normal cell growth (6,7). The tumor suppressor p53 is activated in response to stress such as DNA damage and oncogene activation. p53 can promote cell apoptosis and thus enhance the chemosensitivity to anticancer agents (8,9). Unlike p53, p73 is rarely mutated in human cancers. Due to high homology to p53, the p53-related protein p73 is capable of transactivating p53 target genes and also plays an important role in the regulation of chemosensitivity (10,11). The p53 family members are an attractive pharmaceutical target for cancer therapy. Many compounds have been discovered which can induce the activation of p53 and the death of tumor cells (1215).

4-(3′,3′-Dimethylallyloxy)-5-methyl-6-methoxyphthalide (DMMP) was initially found in the liquid culture of the fungus Alternaria porri (Fig. 1) (16,17). DMMP was reported to have antifungal activity and cytotoxic activity in cancer cell lines. In our research, DMMP was isolated from the plant endophytic fungus Pestalotiopsis photiniae(18). Although the cytotoxic activities of DMMP have been reported, little is known concerning the molecular mechanism of its cytotoxic effect. Here, for the first time, we discovered that DMMP inhibits the growth of several cancer cell lines and we investigated the mechanism of its antiproliferative effect. These results were significant in that they provide a mechanistic framework for further exploration of the use of DMMP as a novel antitumor agent.

Materials and methods

Materials

Caspase inhibitor Z-VAD-FMK, caspase-3 specific inhibitor Z-DEVD-FMK, PD9805 were from Sigma-Aldrich (St. Louis, MO, USA; MTT, acridine orange and ethidium bromide were from Amresco LLC (Solon, OH, USA). JC-1 was from Beyotime Institute of Biotechnology (Jiangsu, China). Rabbit polyclonal antibodies against Fas-L, Fas, p73, p53, Bax, Bcl-2, survivin, CDK2, Akt, p-AKT (Thr308), ERK, p-ERK1/2 (Thr202/Tyr204), p-RB (Ser807/811), GAPDH, Bid, caspase-8, caspase-9, cyclin E, p27KIPI, Bcl-xL and E2F1 were obtained from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA). Mouse monoclonal p53 and mouse monoclonal CDK6 were obtained from Santa Cruz Biotechnology, Inc., and rabbit polyclonal activated caspase-3 was from Cell Signaling Technology, Inc. (Danvers, MA, USA). MEM/NEAA medium, DMEM, L15 medium were from Invitrogen Life Technologies (Carlsbad, CA, USA). Propidium iodide (PI)/RNase staining buffer and the Annexin V-FITC Apoptosis Detection kit were from BD Pharmingen (San Diego, CA, USA). PVDF membranes were from Millipore (Billerica, MA, USA) and the PrimeScript™ High Fidelity RT-PCR kit and SYBR® Premix Dimer Eraser were from Takara Bio, Inc. (Shiga, Japan). DMMP (Fig. 1) was provided by our research group at Hebei University (purity >99%, HPLC analysis) (18).

Cell culture

All cell lines were purchased from Cell Resource Center, IBMS, CAMS/PUMC. MCF7 human breast adenocarcinoma and HeLa human cervical cancer cells were grown in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% heat-inactivated fetal bovine serum (FBS) (Invitrogen Life Technologies). MRC5 normal lung cells were grown in MEM/NEAA supplemented with 10% heat-inactivated FBS. MDA-MB-231 human breast adenocarcinoma cells were grown in L15 medium supplemented with 10% heat-inactivated FBS and 1% glutamine. All cell lines were cultured at 37°C in a humidified incubator containing 5% CO2.

MTT assay

Cell viability was determined using the colorimetric 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. Exponentially growing cells (8,000 cells/well) were seeded in 96-well plates and cultured at 37°C, under 5% CO2 and 95% air for 24 h. Cells were treated with DMMP at different concentrations (5, 10, 20, 40 and 80 μg/ml dissolved in DMSO) for different time periods. The DMSO concentration was maintained below 0.5% which was found to have no antiproliferative effect on the cell lines. Then 20 μl MTT (5 mg/ml) was added to each well. After incubation at 37°C for 4 h, 100 μl 10% SDS-HCl was added and incubated at 37°C overnight. The OD value of the system was measured at a wavelength of 570 nm. Five wells were counted in 3 different experiments. The 50% inhibitory concentration (IC50) of DMMP on cells was calculated by the MTT assay.

Inhibitor treatment

The cell culture was preincubated for 2 h with one of the following inhibitors: the cell-permeable pan-caspase inhibitor Z-VAD-FMK (10 μM), the caspase-3-specific inhibitor Z-DEVD-FMK (10 μM) or ERK inhibitor PD98059 (20 μM) before the addition of DMMP. Cell viability was measured by the MTT assay.

Caspase activity assay

Caspase activation was measured using a caspase colorimetric assay kit (Beyotime Institute of Biotechnology). Briefly, following cell treatment with 40 μg/ml DMMP for the indicated periods of time, cells were harvested and 25 μl of cold lysis buffer/106 cells was added. The cell lysates were incubated on ice for 10 min and then centrifuged at 16,000 × g for 15 min. The protein concentration was determined by Bradford assay. To each reaction well, 10 μl of caspase-3 fluorogenic substrate (Ac-DEVD-pNA) was added to 10 μl sample (1–3 mg/ml) and 80 μl detection buffer. The plate was incubated at 37°C for 2 h. The plate was read on a fluorescence microplate reader.

Acridine orange/ethidium bromide (AO/EB) staining

Exponentially growing cells were seeded on polylysine-coated glass coverslip in a 24-well-plate and cultured at 37°C, under 5% CO2 for 24 h. After incubation with 40 μg/ml DMMP, cells were washed with PBS for 3 times and then stained with 100 μg/ml AO/EB for 5 min. Coverslips were mounted and the fluorescence was visualized using fluorescence microscopy (Olympus).

JC-1 analysis

Cells (104) were seeded on polylysine-coated glass coverslips in a 24-well plate, and incubate at 37°C under 5% CO2 for 24 h. After being exposed to DMMP for 12 and 24 h, cells were washed with PBS for 3 times, and then incubated with a final concentration of 2 μM JC-1 dye at 37°C under 5% CO2 for 20 min. Cells were treated with a final concentration of 50 μM CCCP at 37°C for 20 min as a positive control. After 3 washes with PBS, the coverslips were mounted and the fluorescence was visualized using fluorescence microscopy (Olympus).

Flow cytometric assay

HeLa cells were treated with DMMP at a concentration of 40 μg/ml for 12, 24 and 36 h. Cells (106)were collected by centrifuging at 1,000 rpm for 5 min, and then cells were washed twice with ice-cold PBS. For cell cycle analysis, cells were fixed in ice-cold ethanol (70% v/v) and stained with 0.5 ml PI/RNase staining buffer (BD Pharmingen) for 15 min at room temperature and analyzed by flow cytometry (Becton-Dickinson, Franklin Lakes, NJ, USA). Apoptotic/necrotic cells were detected using the Annexin V-FITC Apoptosis Detection kit (BD Pharmingen). Briefly, cells were incubated with binding buffer (10 mM HEPES/NaOH, pH 7.5, 140 mM NaCl and 2.5 mM CaCl2) and stained with PI and FITC-labeled Annexin V for 15 min at room temperature in the dark. Cell fluorescence was evaluated by flow cytometry using a FACSCalibur instrument and analyzed by Cell Quest software.

Western blotting

Western blotting was used to determine the expression of various key proteins. Cells were seeded in a culture bottle at 37°C under 5% CO2 and 95% air. The cells were treated with DMMP (40 μg/ml for 12, 24 and 36 h). Approximately 2×106 cells were collected and washed with ice-cold PBS. Subsequently, cells were incubated in lysis buffer (50 mM HEPES-NaOH, 100 mM NaCl, 0.5% NP-40, 2.5 mM EDTA, 10% glycerol, 1 mM DTT, 1 mM Na3VO4, 5 mM NaF, 1 mM PMSF, 2 μg/ml pepstatin, 5 μg/ml leupetin, 2 μg/ml aprotintin) for 10 min on ice, collected by scraping and kept on ice for 30 min. Finally, after freezing and thawing, cells were centrifuged at 14,000 rpm for 30 min at 4°C. Cell lysates were centrifuged at 12,000 rpm for 15 min at 4°C. Protein concentrations in lysates were determined by Bradford assay. Fifty micrograms of protein from the protein lysates per sample was denatured in 2X sample buffer and was loaded on sodium dodecyl sulfate-polyacrylamide (SDS) gel electrophoresis on 8–12% Tris-glycine gel. After electrophoresis, proteins were transferred onto PVDF membranes (Millipore) followed by blocking with 5% non-fat milk powder (w/v) in Tris-buffered saline [10 mM Tris-HCl (pH 7.5), 100 mM NaCl, 0.1% Tween-20] for 1 h at room temperature. The membranes were then incubated with specific primary antibodies for 1 h. After washing, the membranes were incubated with appropriate secondary HRP-conjugated antibodies and visualized by ECL. Each membrane was stripped and reprobed with anti-GADPH antibody to ensure equal protein loading.

Real-time RT-PCR

Approximately 106 cells were harvested at the indicated time points and total RNA was isolated using TRIzol reagent (Invitrogen Life Technologies) as described by the manufacturer. The integration of RNA was detected by agarose gel analysis and spectrophotometry. Reverse transcription of total RNA was performed by PrimeScript™ High Fidelity RT-PCR kit (Takara Bio, Inc). Real-time RT-PCR was performed using a Bio-Rad (Hercules, CA, USA) iCycler PCR machine. Each PCR mixture contained 100 ng of cDNA template and primers at a concentration of 10 μM in a final volume of 25 μl of SYBR Premix Dimer Eraser. Specificity of each PCR was examined by the melting temperature profiles of the final products. Standard curves were calculated using cDNA to determine the linear range and efficiency of each primer pair (primer sequences are provided upon request). Reactions were conducted in triplicate, and relative amounts of gene were normalized to GAPDH. The relative gene expression data were analyzed by the comparative CT method (ΔΔCT method).

Statistical analysis

The statistical significance of the differences between the control and treated groups was determined using the 2-tailed Student’s t-test. P-values <0.05 were considered to indicate a statistically significant result.

Results

Effect of DMMP on cell proliferation

To assess the effect of DMMP on cell proliferation, human cancer cell lines, HeLa, MCF7 and MDA-MB-231, and the normal lung fibroblast cell line MRC5 were treated with increasing concentrations of DMMP at 24 h, and cell viability was assessed using the MTT assay (Fig. 2). IC50 for HeLa, MDA-MB-231, MCF7 and MRC5 cells were 36, 51, 81 and 147 μg/ml, respectively. DMMP inhibited the proliferation of the cancer cells in a concentration-dependent manner. DMMP showed the most antiproliferative effect on the HeLa cell line. While the normal lung fibroblast cell line MRC5 exhibited less sensitivity. DMMP exhibited slight selective cytotoxic effect.

Effect of DMMP on cell cycle progression

The effect of DMMP treatment on cell cycle arrest was examined. Compared to DMSO treatment, cells treated with DMMP at the indicated times notably accumulated in the G1 phase of the cell cycle with a reduction in the percentage of cells in S phase (Fig. 3A). DMMP treatment for 24 h increased the percentage of cells in the G1 phase from 68.38 to 92.0%. These results suggest that DMMP inhibited cellular proliferation of HeLa cells via the G1 phase arrest of the cell cycle.

The expression of cell cycle-regulating proteins was examined. Expression levels of cyclin E were gradually increased after DMMP treatment, with slight upregulation of CDK2 (Fig. 3B). p27KIP1 was able to directly bind several different classes of cyclins and CDKs leading to cell cycle arrest in the G1 phase (19). As shown in Fig. 3B, p27KIP1 was dramatically upregulated at 12 h and this effect increased over time. pRb is an important regulator of G1/S phase transition. Phosphorylated pRb was slightly increased (Fig. 3B). E2F1, the binding partner of pRb, was increased at 12 and 24 h.

DMMP induces apoptosis in HeLa cancer cells

To further investigate the mechanism of the inhibitory effect of DMMP, acridine orange/ethidium bromide (AO/EB) staining was used to visualize nuclear changes and apoptotic body formation which are characteristic of apoptosis. AO is a membrane-permeable dye that can pass through the membrane of intact cells and embed in the nuclear DNA emitting bright green fluorescence. EB can pass through the membrane of a dead or dying cell with orange fluorescence. After HeLa cells were exposed to DMMP for 12 h, early apoptotic cells with green pyknotic-like nuclear chromatin were noted. At 24 h, late apoptotic cells with pyknotic-like deep orange nuclear chromatin were detected. Control cells with normal morphology showed green fluorescence (Fig. 4A). The results suggest that DMMP was able to induce marked apoptotic morphology in HeLa cells in a time-dependent manner.

To investigate the kinetics of the apoptotic cells induced by DMMP, HeLa cells were treated with DMMP for various periods of time, and the apoptotic cell death was analyzed by flow cytometry using Annexin V-PI staining. The proportion of early apoptotic cells (Annexin V-positive and PI-negative) was significantly increased from 2.63% in the control cells to 8.52% at 12 h, 38.09% at 24 h, and 44.46% at 36 h following DMMP treatment (Fig. 4B and C). These results indicate that cell death induced by DMMP was mainly caused by induction of apoptosis in a time-dependent-manner.

Caspase activity is induced by DMMP

We investigated whether DMMP induces HeLa cell apoptosis through the caspase pathway. Caspase-8, -9 and -3 were detected by western blotting as shown in Fig. 5A. Following DMMP treatment full-length caspase-9 was decreased in a time-dependent manner. Cleaved caspase-9 was detected indicating activation of caspase-9 and cleaved caspase-3 was also detected, while activated caspase-8 was not detected (Fig. 5A). Meanwhile, expression of the death receptor, Fas, was unchanged and its ligand, FasL was slightly increased (Fig. 5A). Survivin expression was gradually increased and peaked at 24 h and AIF was also slightly increased in DMMP-induced apoptotic cell death (Fig. 5A).

Caspase-3 can be activated either in the intrinsic or the extrinsic apoptotic pathway. To further identify whether caspase-3 is involved in DMMP-induced apoptosis, the catalytic activity of caspase-3 was measured using a caspase colorimetric assay kit. Caspase activity was not significantly increased when compared to the control after DMMP treatment (Fig. 5B). To further evaluate the role of caspases in DMMP-induced apoptosis pathway, HeLa cells were pretreated in the presence of the broad-spectrum caspase inhibitor (Z-VAD-FMK) and caspase-3 inhibitor (Ac-DEVD-CHO) for 2 h before drug treatment. Pretreatment of caspase-3 and broad-spectrum caspase inhibitors failed to attenuate cell death following DMMP treatment (Fig. 5C). These results showed that DMMP-induced apoptosis may be through a caspase-independent mechanism.

DMMP induces loss of mitochondrial transmembrane potential (ΔΨm)

Loss of mitochondrial transmembrane potential is one of several key events that occur in the mitochondria during apoptosis. JC-1 is the most commonly used fluorescent probe to detect the change in mitochondrial membrane potential (ΔΨm). In healthy cells with high ΔΨm, JC-1 spontaneously shows intense red fluorescence and in contrast in apoptotic or unhealthy cells with low ΔΨm, JC-1 shows only green fluorescence. In untreated HeLa cells, JC-1 showed intense red fluorescence. At 12 h after drug treatment, green fluorescence was noted in some cells indicating the dissipation of ΔΨm. At 24 h, most cells exhibited green fluorescence (Fig. 6). The time course analysis revealed that significant numbers of cells following treatment with DMMP lost ΔΨm.

Effect of DMMP on the Bcl-2 family

Bcl-2 family proteins are key regulators of mitochondrial permeability. Therefore, we investigated whether apoptosis in HeLa cells induced by DMMP is modulated by Bcl-2 family members. Western blotting results revealed that pro-survival Bcl-2 family proteins, Bcl-2, was decreased at 36 h, and Bcl-xL was decreased at 12 and 24 h (Fig. 5A). Bcl-2 mRNA expression significantly increased at 12 h (2.5-fold ) and 24 h (2.2-fold), and then decreased at 36 h after treatment consistent with the results of the western blotting. Meanwhile, a pro-apoptotic Bcl-2 family member, Bax, was significantly increased (Fig. 5A). The mRNA expression levels of PUMA, NOXA, Bax, Bad and Bid were significantly upregulated after treatment (Fig. 7). The mRNA expression levels of PUMA and NOXA were significantly increased (3.4- and 5.4-fold, respectively) compared with the control at 12 h and achieved a maximum increase at 36 h after treatment (10.1- and 11.2-fold). Bax and Bid mRNA expression was significantly increased at 24 h after treatment (2.0- and 1.8-fold, respectively). Bim and Bad mRNA expression was significantly increased only at 36 h (3.5- and 3.6-fold, respectively).

Effect of DMMP on p53 and p73 expression

To investigate whether DMMP has an effect on p53 protein expression, we determined the p53 levels in HeLa cells treated with DMMP at the indicated time periods. Western blot analysis showed that DMMP treatment resulted in a time-dependent accumulation of p53 in HeLa cells (Fig. 8). Another p53 family member, p73, was also increased. Moreover, HeLa cells contain HPV E6–E7 genes which promote the degradation of p53 and the retinoblastoma protein. HPV E6–E7 mRNA levels were decreased by 50% following the drug treatment (Fig. 7).

Effect of DMMP on ERK and Akt

Western blot analysis showed that treatment with DMMP led to a slight upregulation of p-ERK in the HeLa cells following treatment while levels of total extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) were not altered (Fig. 9). To further demonstrate the functions of ERK in inducing apoptosis, the effects of the highly specific ERK inhibitor PD98059 were examined. As shown in Fig. 5C, PD98059 significantly increased cell death. This indicates that sustained phosphorylation of ERK inhibits DMMP-induced cell death.

Akt is a serine/threonine kinase involved in cellular survival pathways by inhibiting apoptotic processes. To determine whether Akt is involved in DMMP-induced apoptosis, the phosphorylation status of Akt was examined by western blot analysis. Akt phosphorylation was significantly increased at 12 h and declined at 36 h, while the level of total Akt protein was not altered (Fig. 9).

Discussion

During a search for novel anticancer agents in the culture broth of various endophytic fungi, DMMP was isolated from the endophytic fungus Pestalotiopsis photiniae. In the present study, DMMP inhibited the proliferation of various cancer cell lines and had little effect on normal lung MRC5 cells. As compared with the tumor cell lines analyzed, we found that DMMP had selective cytotoxic effect. Thus, the molecular mechanism of the cytotoxic effect of DMMP was investigated in HeLa cells.

DMMP caused cell cycle arrest in the G1 phase of the cell cycle. DMMP significantly elevated p27KIP1 expression (Fig. 3). p27KIP1 causes cell cycle arrest in G1 phase by binding cyclin E-CDK2 complexes (2022). After DMMP treatment, phospho-Rb was slightly elevated, and E2F1 expression was significantly increased at 12 and 24 h. Meanwhile, CDK2 and cyclin E proteins were also increased (Fig. 3B). The retinoblastoma protein (Rb) exhibits growth inhibitory activity by binding with E2F1. The Rb-E2F1 pathway plays an important role in the regulation of cell cycle progression. Cyclin E-CDK2 can phosphorylate Rb resulting in the activation of E2F-responsive genes (23). Whether the increased levels of CDK2 and cyclin E proteins facilitated E2F function requires further investigation.

DMMP efficiently induced HeLa cell apoptosis characterized by compaction and fragmentation of nuclear chromatin. To further confirm the effects on apoptosis, FITC Annexin V and PI double staining and flow cytometry were performed. Results showed that DMMP induced apoptosis in the HeLa cells. Depletion of ΔΨm in HeLa cells was also detected after treatment. The mitochondrial apoptotic pathway is mainly regulated by Bcl-2 family proteins. Any imbalance in the expression level of pro-apoptotic and anti-apoptotic Bcl-2 members leads to the disruption of the outer mitochondrial membrane (24,25). DMMP significantly upregulated the mRNA levels of pro-apoptotic genes such as Bax, PUMA, NOXA, Bim, Bid and Bad (3- to 11-fold) (Fig. 7). Meanwhile, Bcl-2 and Bcl-xL levels were decreased. Bax protein was significantly increased (Fig. 5A), and Bcl-2 mRNA expression was increased by 2-fold and declined at 36 h. As a result of these changes, the ratios of anti-apoptotic proteins and pro-apoptotic proteins of the Bcl-2 family were significantly reduced during apoptosis. The imbalance led to the loss of ΔΨm after DMMP treatment.

The extrinsic apoptosis pathway is indicated by the ligation of cell surface death receptors to their specific ligands such as Fas/FasL (26). The formation of active caspase-8 is involved in the extrinsic apoptosis pathway. Extrinsic apoptosis in some cells is dependent on the cleavage of Bid. In our study, although FasL was slightly increased, we did not detect the activation of caspase-8 and the truncated form of Bid (Fig. 5A). These results showed that DMMP-induced apoptosis did not occur through the Fas/FasL extrinsic apoptosis pathway.

Sequential activation of caspases plays a central role in the execution-phase of cell apoptosis. As an effector caspase, caspase-3 plays a central role in the extrinsic apoptotic pathway and intrinsic apoptotic pathway. In our study, although cleaved caspase-3 and cleaved caspase-9 were detected by western blotting, no significant activation of caspase-3 proteolytic activity was detected after drug treatment. Survivin, which functions to inhibit caspase activation, was elevated after drug treatment. Elevated survivin may inhibit caspase-3 proteolytic activity. Further experiments showed that pretreatment with caspase inhibitors (Ac-DEVD-CHO and Z-VAD-FMK) failed to attenuate DMMP-induced cell death (Fig. 5C). DMMP induced a slightly higher level of AIF after drug treatment compared with the control (Fig. 5A). Although caspase activation is considered a hallmark of apoptotic cell death, caspase-independent apoptosis has been reported. Apoptosis-induced factors (AIF) and endonuclease G (EndoG) residing in mitochondria are involved in the caspase-independent cell death pathway (27). DMMP may induce apoptosis in a caspase-independent manner. Further experimental investigation is needed for confirmation.

The tumor suppressor transcription factor p53 plays a vital role in cell cycle arrest and apoptosis in response to cellular stress. Western blot analysis showed that DMMP treatment resulted in a significant accumulation of p53 and p73 proteins in HeLa cells (Fig. 8). Normally, p53 protein is rapidly degraded by the ubiquitin-proteome system. Mdm2 is an important negative regulator of the p53 tumor suppressor (28,29). But in HPV-infected cervical cancer cells, HPV E6 is the protein responsible for repressing p53 replacing Mdm2 function (30). In the present study, real-time RT-PCR showed that DMMP caused a ~50% reduction of E6 and E7 mRNA expression when compared to the control (Fig. 7). The dramatic upregulation of p53 protein may be due to the low expression of HPV E6.

In large part, the potent anticancer activity of p53 has been linked to its ability to induce apoptosis through the intrinsic mitochondrial-mediated apoptotic pathway (31). p53 enhances the expression of Bcl-2 family members including Bax, Bid, PUMA and Noxa (32). BH3-only proteins, NOXA and PUMA, are important mediators of p53-induced apoptosis (33). E2F1 is the first member of the E2F transcription factor family. Evidence shows that E2F1 also possesses tumor suppressor functions which can induce apoptosis via p53-dependent and p53-independent pathways (34). Oncogenic signaling by E2F1 has recently been linked to stabilization and activation of the tumor suppressor p53 (35). Puma and Noxa can be regulated by both p53 and E2F1. In our study, E2F1 and p53 were both increased after DMMP treatment. The mRNA levels of Bax, Puma, Noxa and Bid genes were significantly increased (Fig. 7). Whether p53 and E2F1 were involved in DMMP-induced apoptosis requires further investigation using p53 or E2F1 knockdown cells.

Extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) are members of the mitogen-activated protein kinase superfamily that can mediate cell proliferation and apoptosis (36). PhosphoERK1/2 was slightly increased after DMMP treatment (Fig. 8). Treatment with the ERK inhibitor PD98059 and DMMP efficiently induced HeLa cell death. This implied that DMMP combined with PD98059 may represent a novel anticancer strategy. The Akt serine/threonine kinases are critical regulators of cell survival. The activation of the PI3K-Akt pathways promotes tumorigenesis by inhibiting apoptotic processes (37). Following DMMP treatment, phospho-Akt, increased at 12 and 24 h, was slightly detected at 36 h while total Akt was unaltered (Fig. 9). The reason for the elevated phosphorylation of Akt at an early stage requires further investigation.

In conclusion, for the first time we demonstrated that DMMP significantly induced HeLa cell apoptosis and caused cell cycle arrest in G1 phase. We investigated the possible mechanisms involved in DMMP-induced apoptosis. We demonstrated that DMMP-induced apoptosis was through the mitochondrial intrinsic pathway in HeLa cells. DMMP upregulated p27 protein, and caused cell cycle arrest in the G1 phase. The p53 family members, p53 and p73, were increased after drug treatment. Our results indicate that DMMP is a promising cancer therapeutic agent.

Acknowledgements

This study was supported by programs for New Century Excellent Talents in University (NCET-09-0112), the National Natural Science Foundation of China (30901755 and 31171885), the Scientific Research Program of Hebei Provincial Education Bureau (2011107), the Hebei Province Science Fund for Distinguished Young Scholars (C2011201113), and the Program for Changjiang Scholars and Innovative Research Team in the University (IRT1124).

References

1 

Koehn FE and Carter GT: The evolving role of natural products in drug discovery. Nat Rev Drug Discov. 4:206–220. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Miller KI, Qing C, Sze DM and Neilan BA: Investigation of the biosynthetic potential of endophytes in traditional Chinese anticancer herbs. PLoS One. 7:e359532012. View Article : Google Scholar : PubMed/NCBI

3 

Tan RX and Zou WX: Endophytes: a rich source of functional metabolites. Nat Prod Rep. 18:448–459. 2001.PubMed/NCBI

4 

Reed JC: Drug insight: cancer therapy strategies based on restoration of endogenous cell death mechanisms. Nat Clin Pract Oncol. 3:388–398. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Frankfurt OS and Krishan A: Apoptosis-based drug screening and detection of selective toxicity to cancer cells. Anticancer Drugs. 14:555–561. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Chen F, Wang W and El-Deiry WS: Current strategies to target p53 in cancer. Biochem Pharmacol. 80:724–730. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Collavin L, Lunardi A and Del Sal G: p53-family proteins and their regulators: hubs and spokes in tumor suppression. Cell Death Differ. 17:901–911. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Ferreira CG, Tolis C and Giaccone G: p53 and chemosensitivity. Ann Oncol. 10:1011–1021. 1999. View Article : Google Scholar : PubMed/NCBI

9 

Lunghi P, Costanzo A, Mazzera L, Rizzoli V, Levrero M and Bonati A: The p53 family protein p73 provides new insights into cancer chemosensitivity and targeting. Clin Cancer Res. 15:6495–6502. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Bisso A, Collavin L and Del Sal G: p73 as a pharmaceutical target for cancer therapy. Curr Pharm Des. 17:578–590. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Slade N and Horvat A: Targeting p73 - a potential approach in cancer treatment. Curr Pharm Des. 17:591–602. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Lain S, Hollick JJ, Campbell J, et al: Discovery, in vivo activity, and mechanism of action of a small-molecule p53 activator. Cancer Cell. 13:454–463. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Vassilev LT, Vu BT, Graves B, et al: In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science. 303:844–848. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Peirce SK and Findley HW: The MDM2 antagonist nutlin-3 sensitizes p53-null neuroblastoma cells to doxorubicin via E2F1 and TAp73. Int J Oncol. 34:1395–1402. 2009.PubMed/NCBI

15 

Sampath D, Calin GA, Puduvalli VK, et al: Specific activation of microRNA106b enables the p73 apoptotic response in chronic lymphocytic leukemia by targeting the ubiquitin ligase Itch for degradation. Blood. 113:3744–3753. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Phuwapraisirisan P, Rangsan J, Siripong P and Tip-Pyang S: New antitumour fungal metabolites from Alternaria porri. Nat Prod Res. 23:1063–1071. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Suemitsu R, Ohnishi K, Morikawa Y and Nagatomo S: Zinnimidine and 5-(3′,3′-dimethylallyloxy)-7-methoxy-6-methylphthalide from Alternaria porri. Phytochemistry. 38:495–497. 1995.PubMed/NCBI

18 

Yang XL, Zhang S, Hu QB, Luo DQ and Zhang Y: Phthalide derivatives with antifungal activities against the plant pathogens isolated from the liquid culture of Pestalotiopsis photiniae. J Antibiot (Tokyo). 64:723–727. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Malumbres M and Barbacid M: To cycle or not to cycle: a critical decision in cancer. Nat Rev Cancer. 1:222–231. 2001. View Article : Google Scholar : PubMed/NCBI

20 

Hengst L and Reed SI: Translational control of p27KIP1 accumulation during the cell cycle. Science. 271:1861–1864. 1996. View Article : Google Scholar

21 

Pagano M, Tam SW, Theodoras AM, et al: Role of the ubiquitin-proteasome pathway in regulating abundance of the cyclin-dependent kinase inhibitor p27. Science. 269:682–685. 1995. View Article : Google Scholar : PubMed/NCBI

22 

Vervoorts J and Lüscher B: Post-translational regulation of the tumor suppressor p27(KIP1). Cell Mol Life Sci. 65:3255–3264. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Harbour JW and Dean DC: The Rb/E2F pathway: expanding roles and emerging paradigms. Genes Dev. 14:2393–2409. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Danial NN and Korsmeyer SJ: Cell death: critical control points. Cell. 116:205–219. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Chipuk JE and Green DR: How do BCL-2 proteins induce mitochondrial outer membrane permeabilization? Trends Cell Biol. 18:157–164. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Ashkenazi A and Dixit VM: Death receptors: signaling and modulation. Science. 281:1305–1308. 1998. View Article : Google Scholar : PubMed/NCBI

27 

Chipuk JE and Green DR: Do inducers of apoptosis trigger caspase-independent cell death? Nat Rev Mol Cell Biol. 6:268–275. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Brooks CL and Gu W: p53 ubiquitination: Mdm2 and beyond. Mol Cell. 21:307–315. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Sullivan KD, Gallant-Behm CL, Henry RE, Fraikin JL and Espinosa JM: The p53 circuit board. Biochim Biophys Acta. 1825:229–244. 2012.PubMed/NCBI

30 

Diaz D, Santander MA and Chavez JA: HPV-16 E6 and E7 oncogene expression is downregulated as a result of Mdm2 knockdown. Int J Oncol. 41:141–146. 2012.

31 

Chipuk JE and Green DR: Dissecting p53-dependent apoptosis. Cell Death Differ. 13:994–1002. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Hemann MT and Lowe SW: The p53-Bcl-2 connection. Cell Death Differ. 13:1256–1259. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Villunger A, Michalak EM, Coultas L, et al: p53- and drug-induced apoptotic responses mediated by BH3-only proteins puma and noxa. Science. 302:1036–1038. 2003. View Article : Google Scholar : PubMed/NCBI

34 

Wu Z, Zheng S and Yu Q: The E2F family and the role of E2F1 in apoptosis. Int J Biochem Cell Biol. 41:2389–2397. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Stiewe T and Pützer BM: Role of the p53-homologue p73 in E2F1-induced apoptosis. Nat Genet. 26:464–469. 2000. View Article : Google Scholar : PubMed/NCBI

36 

Mebratu Y and Tesfaigzi Y: How ERK1/2 activation controls cell proliferation and cell death: is subcellular localization the answer? Cell Cycle. 8:1168–1175. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Amin AR, Paul RK, Thakur VS and Agarwal ML: A novel role for p73 in the regulation of Akt-Foxo1a-Bim signaling and apoptosis induced by the plant lectin, Concanavalin A. Cancer Res. 67:5617–5621. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October 2013
Volume 30 Issue 4

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
Chen C, Hu S, Luo D, Zhu S and Zhou C: Potential antitumor agent from the endophytic fungus Pestalotiopsis photiniae induces apoptosis via the mitochondrial pathway in HeLa cells. Oncol Rep 30: 1773-1781, 2013
APA
Chen, C., Hu, S., Luo, D., Zhu, S., & Zhou, C. (2013). Potential antitumor agent from the endophytic fungus Pestalotiopsis photiniae induces apoptosis via the mitochondrial pathway in HeLa cells. Oncology Reports, 30, 1773-1781. https://doi.org/10.3892/or.2013.2618
MLA
Chen, C., Hu, S., Luo, D., Zhu, S., Zhou, C."Potential antitumor agent from the endophytic fungus Pestalotiopsis photiniae induces apoptosis via the mitochondrial pathway in HeLa cells". Oncology Reports 30.4 (2013): 1773-1781.
Chicago
Chen, C., Hu, S., Luo, D., Zhu, S., Zhou, C."Potential antitumor agent from the endophytic fungus Pestalotiopsis photiniae induces apoptosis via the mitochondrial pathway in HeLa cells". Oncology Reports 30, no. 4 (2013): 1773-1781. https://doi.org/10.3892/or.2013.2618