Suppression of cancer stem-like phenotypes in NCI-H460 lung cancer cells by vanillin through an Akt-dependent pathway

  • Authors:
    • Songpol Srinual
    • Pithi Chanvorachote
    • Varisa Pongrakhananon
  • View Affiliations

  • Published online on: February 17, 2017     https://doi.org/10.3892/ijo.2017.3879
  • Pages: 1341-1351
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancer stem cells (CSCs) have been reported as a major cause of cancer metastasis and the failure of cancer treatment. Cumulative studies have indicated that protein kinase B (Akt) and its downstream signaling pathway, including CSC markers, play a critical role in the aggressive behavior of this cancer. In this study, we investigated whether vanillin, a major component in Vanilla planifolia seed, could suppress cancer stemness phenotypes and related proteins in the human non-small cell lung cancer NCI‑H460 cell line. A non-toxic concentration of vanillin suppressed spheroid and colony formation, two hallmarks of the cancer stemness phenotype, in vitro in NCI‑H460 cells. Western blot analysis revealed that the CSC markers CD133 and ALDH1A1 and the associated transcription factors, Oct4 and Nanog, were extensively downregulated by vanillin. Vanillin also attenuated the expression and activity of Akt, a transcription regulator upstream of CSCs, an action that was confirmed by treatment with the Akt inhibitor perifosine. Furthermore, the ubiquitination of Akt was elevated in response to vanillin treatment prior to proteasomal degradation. This finding indicates that vanillin can inhibit cancer stem cell-like behavior in NCI‑H460 cells through the induction of Akt-proteasomal degradation and reduction of downstream CSC transcription factors. This inhibitory effect of vanillin may be an alternative approach in the treatment against lung cancer metastasis and its resistance to chemotherapy.

Introduction

Lung cancer has been reported as a major cause of mortality with a high metastasis rate compared with other types of cancers (1). Lung cancer cells contain a small population called tumor-initiating cells or cancer stem cells (CSCs) that have extremely tumorigenic, self-renewal and differentiating properties, resulting in a prolonged tumor status, resistance to chemotherapy and cancer relapse (2,3). Clinical observation has reported that this CSC subpopulation is found in cancer specimens and remains an obstacle for cancer treatment (4). In vitro and in vivo studies have revealed that lung cancer stem cells are extensively resistant to the first-line therapy cisplatin compared with neighboring cancer cells (5,6). These CSCs can maintain the cell survival signaling that provides their strength under severe environments (7). Therefore, the attenuation or removal of CSCs would be likely to improve patient outcome.

Like other types of stem cells, CSCs express specific surface markers, including octamer-binding transcription factor 4 (Oct4), Nanog, ATP-binding cassette subfamily G member 2 (ABCG2), and CD133. Oct4 and Nanog are transcription factors that are responsible for maintaining pluripotency, self-renewal proliferation and tumorigenicity in both normal stem cells and cancer stem cells (8,9). Several studies have reported that an elevation of Oct4 and Nanog is tightly related to a low survival rate and high incidence of cancer metastases (10,11). In lung cancer, both Oct4 and Nanog are required to maintain the CSC-like phenotype (12). Overexpression of Oct4 and Nanog enhances spheroid formation in vitro and significantly increases new tumor formation in vivo. Furthermore, CD133-positive lung cancer cells isolated from patients exhibited a high level of Oct4 and ABCG2, and displayed a clear resistance to chemotherapy and radiotherapy (13). Conversely, the attenuation of Oct4 and Nanog expression levels using RNA interference in CSCs led to the loss of the ability to form spheroids and enhanced the sensitivity of these cells to chemotherapy (12,14).

Cumulative research has identified the mechanism regulating cancer stemness properties. Recently, the Akt pathway was demonstrated to be an upstream signaling pathway of Oct4, which is linked to the CSC-like phenotype (15). The phosphorylation of Akt promotes stemness through the upregulation of both the mRNA and protein levels of Oct4 and Nanog (15,16). Akt-positive cancers that express CSC markers can establish colonies in vitro and in vivo (17,18). In contrast, Akt-knockdown experiments using RNA silencing resulted in an extensive suppression of Oct4 as well as an impeded stemness behavior (19). The alteration of both Akt activity and expression may disrupt Oct4 and Nanog functions, and consequently, the CSC properties of cancer.

Several strategies to the suppression of the CSC mechanism have been intensively emphasized. Natural substances from plant sources, including vanillin, have gained increased interest in an alternative therapy due to their various pharmacological activities (20). Vanillin, 4-hydroxy-3-methoxybenzaldehyde (Fig. 1), is the main active ingredient found in the Vanilla planifolia seed. It is widely used as a flavoring agent in many products including food and cosmetics (21). Previous reports have demonstrated several biological activities of vanillin such as antimicrobial, hypotriglyceridemic, anti-inflammatory and antimutagenic activities in rodents and humans (2224). Vanillin has been shown to inhibit cancer invasion and migration through the reduction of matrix metalloprotease activity and downregulation of nuclear factor-κB in hepatocellular carcinoma cells (25). Vanillin could also attenuate the formation of lamellipodia and angiogenesis in lung cancer via the suppression of PI3K activity and inducing the apoptosis of various cancer types, such as human cervical cancer and breast cancer (2628). However, the pharmacological effect of vanillin and its mechanism on CSC-like properties in lung cancer has not been elucidated. In this study, we investigated the effects of vanillin on the CSC phenotypes in the non-small cell lung cancer (NSCLC) NCI-H460 cell line. We herein demonstrated for the first time that vanillin can suppress the stemness behavior of NCI-H460 cells, including spheroid formation, with a reduction in CSC markers. Furthermore, our data showed that Akt is the primary target of the vanillin-attenuating CSC behavior, suggesting that vanillin exhibits potential for further anticancer development.

Materials and methods

Cells and culture conditions

The human NSCLC cell line NCI-H460 was purchased from the American Type Culture Collection, ATCC (Manassas, VA, USA) and was cultured as monolayers in Roswell Park Memorial Institute-1640 medium (RPMI) supplemented with 10% fetal bovine serum (FBS), 2 mM L-glutamine and 100 U/ml penicillin and streptomycin. Cell cultures were incubated at 37°C in a humidified incubator filled with 5% CO2. Cells were subcultured routinely with 0.25% (w/v) trypsin in 0.53 mM ethylenediamine tetraacetic acid (EDTA) and were seeded according to the manufacturer's instructions at ~70% confluence.

Reagents and antibodies

Vanillin (Fig. 1), as USP-secondary standard purity grade (>99.0% purity) was purchased from Sigma-Aldrich (St. Louis, MO, USA). The vanillin stock solution was freshly diluted with RPMI medium, and the treatment solution was prepared by dilution of the stock solution with culture media to the desired concentrations. Cell culture media were also used as the control solvent for the experiments. Glutamate, penicillin-streptomycin antibiotics, and phosphate-buffered saline (PBS) were purchased from Gibco (Grand Island, NY, USA). Methanol, dimethyl sulfoxide (DMSO) and mouse monoclonal antibodies to Oct4 (1:1,000) and ubiquitin (1:1,000) were purchased from Sigma-Aldrich. Rabbit monoclonal antibodies to ABCG2 (1:1,000), β-catenin (1:1,000), Nanog (1:1,000) and phosphorylated Akt (P-Akt, (1:1,000) in Ser473 and Thr308 were from Cell Signaling Technology, Inc. (MA, USA). A goat monoclonal antibody against ALDH1A1 (1:1,000) was purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA), and rabbit monoclonal antibody against CD133 (1:1,000) was purchased from United States Biological (MA, USA).

Cell viability and cell proliferation assays

Cell viability and proliferation assays were performed using the surrogate 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) colorimetric assay. Briefly, NCI-H460 cells were seeded onto 96-well plates at 10,000 cells/well and 2,500 cells/well for the cell viability and proliferation assays, respectively. After cell attachment, various concentrations of vanillin (0–400 µM) were added for 24 h for the cell viability assay. Cells pretreated with the same concentration of vanillin for 1 and 3 days were subcultured and incubated for 24, 48 and 72 h for the cell proliferation assays. At the end of each incubation time, the medium was removed and replaced with MTT solution (Life Technologies, Carlsbad, CA, USA) and incubated for 3 h at 37°C. The MTT solution was then substituted with 100 µl of DMSO to dissolve the formazan crystals, and then the absorbance was measured at 570 nm using a microplate reader. The absorbance was calculated and represented as the % viability and % relative proliferation, respectively, compared with the untreated (control) cells that were set at 100%.

Determination of apoptotic and necrotic cell death

Hoechst 33342 and propidium iodide (PI) (Sigma-Aldrich) were used to stain the nuclei to identify necrotic and apoptotic cell death, respectively. Cells were treated with vanillin for 24 h and then were washed with PBS, followed by incubation with either 5 µM PI or 10 µM Hoechst 33342 at 37°C for 30 min. Cells were visualized and captured under a fluorescence microscope at ×10 magnification (Olympus IX5; 10X with DP70 digital camera system; Olympus, Tokyo, Japan). PI-positive necrotic cells and nuclear condensation of apoptotic cells were scored and reported as the % of all cells viewed.

Anchorage-independent growth assay

The NCI-H460 cells were pretreated with vanillin (0–50 µM) for 1 and 3 days before being subjected to the soft agar colony-formation assay to determine the anchorage-independent growth property of cancer stemness. Soft agar was prepared using a combination of complete media and 1% (w/v) agarose at a 1:1 ratio. This mixture was allowed to solidify in 24-well plates as a bottom layer. Next, pretreated cells suspended in RPMI complete media were mixed with 0.3% (w/v) agarose, added onto the prepared bottom layer and then incubated at 37°C. Further complete media were applied every 2 days to prevent the soft agar drying. After 2 weeks, the colony was examined under a phase-contrast microscope at ×4 magnification (Olympus IX5; 4X with DP70 digital camera system; Olympus). The relative colony number and size were analyzed compared with those of the control group.

Spheroid formation assay

Vanillin-pretreated cells at a cell density of 2,500 cells/well were cultured in 24-well ultralow attachment plates in RPMI serum-free medium for 7 days. Next, the spheroids were disaggregated by trypsinization, resuspended in RPMI serum-free medium as a single cell suspension and then cultured onto 24-well ultralow attachment plates. After an additional 14 days, the presence of secondary spheroids was investigated by phase-contrast microscopy at ×4 magnification (Olympus 1X51 with DP70). The relative spheroid number and size were analyzed compared with those of the control group.

Western blot analysis

Cells were lysed using lysis buffer containing 20 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1% Triton X-100, 1 mM Na3VO4, 10% glycerol, 50 mM NaF, 100 mM phenylmethylsulfonyl fluoride (PMSF; Sigma-Aldrich), and a protease inhibitor cocktail (Merck Millipore, MA, USA), sonicated and incubated on an ice bath for 45 min. The protein content was measured using the BCA protein assay kit (Pierce™ Thermo Fisher Scientific Inc., IL, USA). An equal amount of denatured protein was separated by electrophoresis [10% (w/v) acrylamide resolving gel] and transferred to a 0.45-µm nitrocellulose membrane (Bio-Rad, Hercules, CA, USA). Each membrane was blocked with 5% (w/v) nonfat-milk in Tris-buffered saline solution containing 1% (v/v) Tween-20 (TBST) for at least 30 min and then incubated with the indicated primary antibody at 4°C overnight. Thereafter, the membranes were washed three times with TBST and then incubated with secondary antibody at room temperature for 2 h. The antigen-antibody complexes were detected, after washing with TBST as above, using chemiluminescent solution (Pierce Biotechnology) and were exposed to film (Carestream Health, Inc., Rochester, NY, USA). The densitometry of the target protein was measured using the NIH ImageJ program and quantified as the relative expression level to that of the control group.

Immunoprecipitation assay

Protein interaction was examined by immunoprecipitation. Cells were incubated with 10 µM lactacystin for 1 h prior to treatment with vanillin to inhibit proteasome function while preserving the ubiquitin-protein complex. Treated cells were collected and lysed in TMN buffer (50 mM Tris-HCl pH 7.5, 140 mM NaCl and 0.5 mM MgCl2) containing 10% glycerol, a protease inhibitor cocktail, 1% nonylphenylpolyethylene glycol (NP-40) and 1% PMSF for 30 min, followed by centrifugation at 12,000 rpm at 4°C for 15 min. The supernatants were blocked with protein G agarose beads (GE Healthcare, Little Chalfont, UK) to remove non-specific binding. After centrifugation at 3,000 rpm at 4°C for 5 min, the protein contents were measured. The supernatants were collected and stored at 4°C as an input for immunoblotting where 300 µg of protein was incubated with anti-Akt antibody at 4°C overnight and then incubated with protein G agarose beads for 2 h at 4°C. The immunoprecipitates were collected, washed with TMN buffer and resuspended in 30 µl of 2X SDS sample buffer. The samples were boiled at 95°C for 5 min and then were subjected to western blot analysis using antibodies against ubiquitin.

Statistical analysis

The data were collected from at least four independent experiments, normalized by the control groups and presented as the means ± standard deviation (SD). The significant difference among the groups was analyzed using one-way ANOVA followed by the post hoc test. Statistical analysis was performed using SPSS software (IBM Inc., NY, USA) and statistical significance was accepted at the p<0.05 level.

Results

Cytotoxicity and anti-proliferative effect of vanillin on NCI-H460 cells

To diminish the interference of the cytotoxic effect of vanillin on cancer stemness, a non-toxic concentration of vanillin was used on the NCI-H460 cells, as determined by the cytotoxicity assay. Cells were treated with various concentrations of vanillin for 24 h and were then subjected to the MTT cell viability assay. Fig. 2A demonstrates that 200 µM vanillin caused a significant (p=0.003) mortality with ~30% cell death, whereas at a concentration of <100 µM at least 80% viable cells remained. To confirm the cell death mechanism, cells were similarly treated with vanillin for 24 h and then were incubated with either Hoechst 33342 or PI, where apoptotic nuclei and necrotic bodies were clearly observed in the cells treated with a high (200 µM) dose of vanillin (Fig. 2B and C). This result is consistent with the cell viability testing, revealing that vanillin at concentrations of <100 µM are non-toxic to NCI-H460 cells, so these doses were used for subsequent experiments.

The effect of vanillin on NCI-H460 cell proliferation was also investigated. The cells were pre-treated with vanillin for 1 and 3 days, prior to measuring their proliferation after a further 24, 48 and 72 h. The proliferation of the cells pretreated with 100 µM vanillin for 3 days was significantly reduced as early as 24 h, whereas lower doses (<100 µM) had no significant effect at any detection time (Fig. 2E). Furthermore, pretreatment with vanillin for 1 day showed non-detectable changes compared with that in the control group (Fig. 2D). These results indicated that a low dose of vanillin (0–50 µM) neither induced cell death nor inhibited cell proliferation in the NCI-H460 cells. Cell growth and survival in the absence of extracellular matrix is one of the crucial behaviors of the cancer stem-like phenotype. Thus, to avoid any anti-proliferative effect of vanillin on cancer stem cell growth, vanillin doses that showed no such effect on NCI-H460 cells under the attachment condition were then used for the cancer stem-like phenotype characterization.

Inhibitory effect of vanillin on the anchorage-independent growth and spheroid formation of NCI-H460 cells

Because self-renewal and tumorigenicity are distinctive behaviors for the cancer stem-like phenotype, anchorage-independent growth and spheroid formation have been used to characterize these features. To investigate the suppressive effect of vanillin on colony and spheroid formation, NCI-H460 cells were pretreated with vanillin (0–50 µM) for 1 and 3 days, followed by examination of their anchorage-independent growth and spheroid formation. Fig. 3A shows representative images in which both the colony size and number of cells pretreated with vanillin for 1 day were not different from those of untreated control cells. However, a significant and vanillin dose-dependent reduction in the cell size and number was observed when pre-incubated with vanillin for 3 days (Fig. 3B and C). Approximately 80 and 90% reduction in the colony size and number, respectively, were observed in the NCI-H460 cells pretreated with 50 µM vanillin for 3 days (p=0.027).

The effect of vanillin on the self-renewal property was evaluated by the spheroid formation assay. Primary spheroids, consisting of several types of cells, including progenitors, mature cells and stem cells (29), were first initiated for 7 days and then dissociated to single cells. The single cells were allowed to grow under detachment conditions to form secondary spheroids over 14 days, reflecting the pluripotency of CSCs. Interestingly, pretreatment with vanillin for 3 days markedly attenuated the number of secondary spheres formed under detachment conditions, with only ~0.2-fold of the number of sphere remaining after pretreatment with 50 µM vanillin for 3 days (p=0.022). In contrast, this effect was not found with a 1-day pretreatment with vanillin (Fig. 3D–F). These results suggest that vanillin can abolish the cancer stem-like phenotype of CSCs, in terms of both spheroid formation and anchorage-independent growth.

Vanillin downregulates stemness markers in NCI-H460 cells

Cancer stem cell markers are differentially expressed in different types of cancer, with CD133, ABCG2 and ALDH1A1 being distinctly expressed in lung cancer stem cells (3). Given the inhibitory effect of vanillin on the CSC phenotype, its effect on stemness cell markers was then evaluated in the NCI-H460 cells. NCI-H460 cells were incubated with vanillin (0–50 µM) for 1 and 3 days, and then the expression of CSCs markers was determined. Based on western blot analysis (Fig. 4), CSC protein marker expression was not significantly changed in response to the treatment with vanillin for 1 day (50 µM, p=0.124). However, the level of CSC markers, including those of CD133, ABCG2 and ALDH1A1, were markedly attenuated in a dose-dependent manner after treatment with vanillin for 3 days (Fig. 4B). Quantitative analysis demonstrated that a reduction in the expression level of these proteins was observed in cells treated with vanillin for 3 days at concentrations as low as 10 µM (p=0.021). Thus, vanillin suppresses not only the CSC phenotype but also CSC markers in NCI-H460 cells.

Vanillin suppression of Nanog and Oct4 expression is mediated by an Akt-dependent mechanism

The CSC phenotypes include not only an elevated expression of stemness markers, but also high levels of the related transcription factors, including Nanog and Oct4 (1012) that endow a self-renewal property to the cells (30). To assess whether the negative regulation of CSCs properties by vanillin involved these transcription factors, NCI-H460 cells were treated with various concentrations of vanillin for 1 and 3 days, and then protein expression levels of Oct4 and Nanog were evaluated. As shown in Fig. 4B, the Oct4 and Nanog protein expression levels were significantly decreased in the cells treated with vanillin for 3 days, but not in the cells treated for 1 day (Fig. 4A). These results illustrate a good correlation with the other stemness phenotypes (Fig. 3 and 4), demonstrated by their suppression after treatment with vanillin for 3 days.

Because the expression levels of Nanog and Oct4, which contribute to the stemness behavior, are regulated directly by Akt (15), whether vanillin modulates Akt protein expression levels or its activity in terms of P-Akt expression was evaluated next. NCI-H460 cells were treated with vanillin for 3 days prior to determination of the Akt and P-Akt expression levels. Western blot analysis showed that the level of P-Akt (Ser473 and Thr308) or active Akt, was decreased (Fig. 5A), suggesting that the reduced CSCs phenotypes may result from an Akt-dependent mechanism. To verify whether the reduction of P-Akt by vanillin is a result of the suppression of P-Akt or total Akt expression levels, the total Akt expression level was also investigated. As shown in Fig. 5A, the total Akt level was clearly downregulated in a similar pattern. Densitometry analysis of P-Akt relative to total Akt was then analyzed to confirm the hypothesis that vanillin interferes with the Akt expression level. The quantitative results demonstrated that the ratio of P-Akt over its parental form are also changed, indicating the reduction of Akt expression may be, at least in part, a mechanism by which vanillin suppresses Akt activity.

To confirm the above finding that the downregulation of Nanog and Oct4 by vanillin is by an Akt-dependent pathway, perifosine (1,1-dimethylpiperidinium-4-yl octadecyl phosphate), an Akt inhibitor, was used as a positive control. H460 cells were treated with perifosine (0–5 µM) for 3 days, and the mentioned proteins were observed by western blotting as before. Interestingly, inactivation of Akt by perifosine caused a reduction of its downstream signaling, including Oct4 and Nanog (Fig. 5B), similar to that with vanillin (Fig. 4B). The CSC markers CD133, ABCG2 and ALDH1A1 were also downregulated corresponding to the suppression of Akt activity. Although the vanillin-blocked Akt activation occurs partly through downregulating the parental form, the total effect is similar to that by perifosine. These data indicated the Akt function that promotes cancer stemness property via Oct4 and Nanog, which was conversely inhibited by vanillin.

Vanillin mediates Akt degradation through the ubiquitin-proteasomal pathway

Emerging evidence has indicated that the existence of Akt is mainly governed by ubiquitin-proteasomal degradation via E3 ligase (31,32). The reduction of Akt by the enhancement of this mechanism results in the interference of its downstream signaling mediators, including Oct4 and Nanog (11,15,16,33,34). Accordingly, we next examined whether downregulation of Akt by vanillin occurred through the ubiquitin proteasomal pathway. Protein degradation by this pathway requires the protein to bind with ubiquitin prior to recognition by the proteasome (35). NCI-H460 cells were pretreated with 10 µM lactacystin (Lac), a proteasomal inhibitor, prior to vanillin treatment (50 µM) for 3 h, and the presence of Akt-ubiquitin complex was determined by immunoprecipitation. Fig. 5C shows that the level of the Akt-ubiquitin interaction was markedly increased ~2-fold in the vanillin-treated group, which indicated that suppression of CSCs by vanillin may be a consequence of vanillin promoting Akt degradation through the ubiquitin-proteasomal pathway.

Discussion

Taken together, cancers are one of the most common non-accident related cause of deaths among human populations, but the successful rate of treatment is low due to the resistance to chemotherapy treatment, metastasis and the self-renewal properties of cancer cells. It has been reported that these aggressive behaviors are driven by CSCs (2), which are a small population found within tumors, especially in metastatic cancers (3). These CSCs exhibit an overexpression of specific markers such as CD133, ALDH1A1 and ABCG2 (2,36), and related proteins such as Akt and mTOR (37). Recently, research into drug discovery and development for cancer therapy has focused on exploring new natural compounds that target CSCs and related pathways. Several reports have suggested that many natural substances, such as curcumin, resveratrol and pomegranate extract, have the ability to inhibit CSCs (3840).

Herein, we report for the first time that vanillin, an active component in Vanilla planifolia, suppresses CSCs in term of both their phenotype and related molecular mechanisms. A non-cytotoxic concentration of vanillin (50 µM) attenuated spheroid formation and anchorage-independent growth, which are the two prominent characteristics of CSCs (Fig. 3). The inhibitory effects of vanillin were clearly observed after treatment for 3 days, suggesting that in the early phase of treatment, a compensation mechanism by other kinase proteins, such as mitogen activated protein kinase (MAPK) or ERK may exist (41,42). Both PI3K/Akt and MAPK/ERK are known to play an important role in the maintenance of cell survival, differentiation and proliferation of cancer (43,44). A recent study reported that epidermal growth factor (EGF) could induce Akt activation through the MEK inhibitor (45). This phenomenon may occur during the 1-day treatment course of vanillin. An alternative possibility is that cellular signaling requires a longer activation time to induce the phenotypic changes. For example, the inhibitory effect of gigantol on CSCs was found only after treatment for at least 2 days (46). The ability of cancer cells to form neurospheres also indicates the pluripotency of CSCs (45), and our finding here demonstrated that vanillin greatly suppresses the in vitro formation of secondary spheroids consisting of NCI-H460 cells (Fig. 3D).

CD133, ALDH1A1 and ABCG2 have been reported to be specific markers in lung CSCs both in vivo and in vitro (4749). Lung CSCs with CD133+ implanted in mice exhibited a self-renewal property, chemoresistance and a high tumorigenic capability. Like the in vitro study, NCI-H460 and NCI-A549, which express high levels of ABCG2 and CD133, can form spheroids, but this ability was attenuated after reduction of ABCG2 and CD133 expression (50). We further evaluated the expression of such markers in vanillin-treated cells. Corresponding to the phenotypic changes, vanillin downregulated these lung CSC markers (Fig. 4). As transcription factors, Oct4 and Nanog are frequently found in both CSCs and normal stem cells (49), and are responsible for maintaining the self-renewal and pluripotent properties of CSCs under regulation by the kinase activity of Akt (51). These transcription factors are upregulated in many cancers, especially lung cancer, and the reduction of Oct4 and Nanog expression can directly inhibit CSC phenotypes (11,30). Knockdown of these transcription factors was shown to decrease the proliferation and spheroid formation of cancer cells (15). Supporting our finding, Oct4 and Nanog were extensively decreased in response to vanillin treatment, and so likely contributed to the impedance of neurosphere formation and colony-formation capability of the NCI-H460 cells (Fig. 3).

Evidence has suggested that Akt plays a vital role in cell survival and proliferation (52), and drive cancer cells to CSC-like phenotypes including proliferation, migration and self-renewal properties, through the action of the transcription factors Oct4 and Nanog (16). Blockade of Akt was shown to inhibit the in vitro proliferation of spheroid formation and enhanced Oct4 degradation (33). In breast cancer, inhibition of PI3K or Akt activity led to pluripotency loss (37). Furthermore, in prostate cancer, the alteration of the PTEN/PI3K/Akt pathway interfered with the CSC phenotype (53). Likewise, in this study, vanillin was found to diminish Akt function and its expression, together with a decreased CSC behavior and marker expression (Fig. 4). As a key protein catabolism pathway, the ubiquitin-proteasomal pathway is known to regulate several cellular signaling molecules, including Akt (54). Targeted proteins for degradation are poly-ubiquitinated by the ubiquitin ligase (E3) of the proteasome prior to recognition by ubiquitin-activating E1 and E2. The results of this study are consistent with the notion that vanillin drives Akt degradation through elevation of the ubiquitin-proteasomal pathway (Fig. 5). However, the mechanism that leads to ubiquitinized Akt is still unknown. Recent studies reported that multiple growth factor receptors such as EGFR and insulin-like growth factor 1 receptor, might be possible targets of natural compounds (55,56). Since the ubiquitination-proteasomal degradation of intracellular proteins is modulated by the downstream signaling of these receptors (5760), it is possible that the mechanism of vanillin action on ubiquitinized Akt might involve vanillin-modulating of these receptors and their signaling pathways. However, the ubiquitin-proteasomal pathway is also regulated by the reactive oxygen species (61,62) and the exogenous compounds, that alter the oxidative status of the cells, affect this degradation pathway (63,64). Since vanillin has been demonstrated to have a dual effect as an oxidative stress scavenging and pro-oxidant activity, depending on the cell type and vanillin concentration (6567), thus, it is also possible that vanillin may promote Akt degradation through this mechanism.

In conclusion, this study demonstrates that vanillin can suppress the CSC-like phenotype in NCI-H460 cells via the enhancement of the Akt-ubiquitin proteasome degradation pathway, resulting in the downregulation of the transcription factors Oct4 and Nanog (Fig. 6). This finding may provide a new strategy to overcome CSCs in lung cancer.

Abbreviations:

ABCG2

ATP-binding cassette subfamily G member 2

ALDH1A1

aldehyde dehydrogenase 1 family member A1

Akt

protein kinase B

CSCs

cancer stem cells

DMSO

dimethyl sulfoxide

MTT

3-(4,5-dimethylthiazol-2-Yl)-2,5-diphenyl tetrazolium bromide

EDTA

ethylenediamine tetraacetic acid

FBS

fetal bovine serum

NSCLC

non-small cell lung cancer

Oct4

octamer-binding transcription factor 4

P-Akt

active Akt (phosphorylated at Ser473 and Thr308 residues)

PBS

phosphate-buffered saline

PI

propidium iodide

PMSF

phenylmethylsulfonyl fluoride

RPMI

Roswell Park Memorial Institute

TBST

Tris-buffered saline solution with 1% Tween-20

Acknowledgments

This study was supported by grant for International Research Integration: Chula Research Scholar, Ratchadaphiseksomphot Endowment Fund, and the 90th Anniversary Chulalongkorn University. The English editing was proofed by The American Journal Expert service.

References

1 

Peters S, Adjei AA, Gridelli C, Reck M and Kerr K: Metastatic non-small-cell lung cancer (NSCLC): ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 23(Suppl 7): vii56–vii64. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Dalerba P, Cho RW and Clarke MF: Cancer stem cells: Models and concepts. Annu Rev Med. 58:267–284. 2007. View Article : Google Scholar

3 

Medema JP: Cancer stem cells: The challenges ahead. Nat Cell Biol. 15:338–344. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Pallini R, Ricci-Vitiani L, Banna GL, Signore M, Lombardi D, Todaro M, Stassi G, Martini M, Maira G, Larocca LM, et al: Cancer stem cell analysis and clinical outcome in patients with glioblastoma multiforme. Clin Cancer Res. 14:8205–8212. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Lopez-Ayllon BD, Moncho-Amor V, Abarrategi A, Ibañez de Cáceres I, Castro-Carpeño J, Belda-Iniesta C, Perona R and Sastre L: Cancer stem cells and cisplatin-resistant cells isolated from non-small-lung cancer cell lines constitute related cell populations. Cancer Med. 3:1099–1111. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Yang J, Guo W, Wang L, Yu L, Mei H, Fang S, Ji P, Liu Y, Liu G and Song Q: Cisplatin-resistant osteosarcoma cells possess cancer stem cell properties in a mouse model. Oncol Lett. 12:2599–2605. 2016.PubMed/NCBI

7 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Yin X, Zhang BH, Zheng SS, Gao DM, Qiu SJ, Wu WZ and Ren ZG: Coexpression of gene Oct4 and Nanog initiates stem cell characteristics in hepatocellular carcinoma and promotes epithelial-mesenchymal transition through activation of Stat3/Snail signaling. J Hematol Oncol. 8:232015. View Article : Google Scholar : PubMed/NCBI

9 

Loh YH, Wu Q, Chew JL, Vega VB, Zhang W, Chen X, Bourque G, George J, Leong B, Liu J, et al: The Oct4 and Nanog transcription network regulates pluripotency in mouse embryonic stem cells. Nat Genet. 38:431–440. 2006. View Article : Google Scholar : PubMed/NCBI

10 

He W, Li K, Wang F, Qin YR and Fan QX: Expression of OCT4 in human esophageal squamous cell carcinoma is significantly associated with poorer prognosis. World J Gastroenterol. 18:712–719. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Lu X, Mazur SJ, Lin T, Appella E and Xu Y: The pluripotency factor nanog promotes breast cancer tumorigenesis and metastasis. Oncogene. 33:2655–2664. 2014. View Article : Google Scholar :

12 

Chiou SH, Wang ML, Chou YT, Chen CJ, Hong CF, Hsieh WJ, Chang HT, Chen YS, Lin TW, Hsu HS, et al: Coexpression of Oct4 and Nanog enhances malignancy in lung adenocarcinoma by inducing cancer stem cell-like properties and epithelial-mesenchymal transdifferentiation. Cancer Res. 70:10433–10444. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Chen YC, Hsu HS, Chen YW, Tsai TH, How CK, Wang CY, Hung SC, Chang YL, Tsai ML, Lee YY, et al: Oct-4 expression maintained cancer stem-like properties in lung cancer-derived CD133-positive cells. PLoS One. 3:e26372008. View Article : Google Scholar : PubMed/NCBI

14 

Lu Y, Zhu H, Shan H, Lu J, Chang X, Li X, Lu J, Fan X, Zhu S, Wang Y, et al: Knockdown of Oct4 and Nanog expression inhibits the stemness of pancreatic cancer cells. Cancer Lett. 340:113–123. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Zhao QW, Zhou YW, Li WX, Kang B, Zhang XQ, Yang Y, Cheng J, Yin SY, Tong Y, He JQ, et al: Akt-mediated phosphorylation of Oct4 is associated with the proliferation of stem-like cancer cells. Oncol Rep. 33:1621–1629. 2015.PubMed/NCBI

16 

Lin Y, Yang Y, Li W, Chen Q, Li J, Pan X, Zhou L, Liu C, Chen C, He J, et al: Reciprocal regulation of Akt and Oct4 promotes the self-renewal and survival of embryonal carcinoma cells. Mol Cell. 48:627–640. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Bleau AM, Hambardzumyan D, Ozawa T, Fomchenko EI, Huse JT, Brennan CW and Holland EC: PTEN/PI3K/Akt pathway regulates the side population phenotype and ABCG2 activity in glioma tumor stem-like cells. Cell Stem Cell. 4:226–235. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Wang YK, Zhu YL, Qiu FM, Zhang T, Chen ZG, Zheng S and Huang J: Activation of Akt and MAPK pathways enhances the tumorigenicity of CD133+ primary colon cancer cells. Carcinogenesis. 31:1376–1380. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Noh KH, Kim BW, Song KH, Cho H, Lee YH, Kim JH, Chung JY, Kim JH, Hewitt SM, Seong SY, et al: Nanog signaling in cancer promotes stem-like phenotype and immune evasion. J Clin Invest. 122:4077–4093. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Rajesh E, Sankari LS, Malathi L and Krupaa JR: Naturally occurring products in cancer therapy. J Pharm Bioallied Sci. 7(Suppl 1): S181–S183. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Walton NJ, Mayer MJ and Narbad A: Vanillin. Phytochemistry. 63:505–515. 2003. View Article : Google Scholar : PubMed/NCBI

22 

King AA, Shaughnessy DT, Mure K, Leszczynska J, Ward WO, Umbach DM, Xu Z, Ducharme D, Taylor JA, Demarini DM, et al: Antimutagenicity of cinnamaldehyde and vanillin in human cells: Global gene expression and possible role of DNA damage and repair. Mutat Res. 616:60–69. 2007. View Article : Google Scholar :

23 

Srinivasan K, Platel K and Rao MVL: Hypotriglyceridemic effect of dietary vanillin in experimental rats. Eur Food Res Technol. 228:103–108. 2008. View Article : Google Scholar

24 

Imanishi H, Sasaki YF, Matsumoto K, Watanabe M, Ohta T, Shirasu Y and Tutikawa K: Suppression of 6-TG-resistant mutations in V79 cells and recessive spot formations in mice by vanillin. Mutat Res. 243:151–158. 1990. View Article : Google Scholar : PubMed/NCBI

25 

Liang JA, Wu SL, Lo HY, Hsiang CY and Ho TY: Vanillin inhibits matrix metalloproteinase-9 expression through downregulation of nuclear factor-kappaB signaling pathway in human hepatocellular carcinoma cells. Mol Pharmacol. 75:151–157. 2009. View Article : Google Scholar

26 

Lirdprapamongkol K, Sakurai H, Suzuki S, Koizumi K, Prangsaengtong O, Viriyaroj A, Ruchirawat S, Svasti J and Saiki I: Vanillin enhances TRAIL-induced apoptosis in cancer cells through inhibition of NF-kappaB activation. In Vivo. 24:501–506. 2010.PubMed/NCBI

27 

Lirdprapamongkol K, Sakurai H, Kawasaki N, Choo MK, Saitoh Y, Aozuka Y, Singhirunnusorn P, Ruchirawat S, Svasti J and Saiki I: Vanillin suppresses in vitro invasion and in vivo metastasis of mouse breast cancer cells. Eur J Pharm Sci. 25:57–65. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Lirdprapamongkol K, Kramb JP, Suthiphongchai T, Surarit R, Srisomsap C, Dannhardt G and Svasti J: Vanillin suppresses metastatic potential of human cancer cells through PI3K inhibition and decreases angiogenesis in vivo. J Agric Food Chem. 57:3055–3063. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Bachelard-Cascales E, Chapellier M, Delay E and Maguer-Satta V: A protocol to quantify mammary early common progenitors from long-term mammosphere culture. Curr Protoc Stem Cell Biol. 1:p Unit 1E. 72012.PubMed/NCBI

30 

Amini S, Fathi F, Mobalegi J, Sofimajidpour H and Ghadimi T: The expressions of stem cell markers: Oct4, Nanog, Sox2, nucleostemin, Bmi, Zfx, Tcl1, Tbx3, Dppa4, and Esrrb in bladder, colon, and prostate cancer, and certain cancer cell lines. Anat Cell Biol. 47:1–11. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Bae S, Kim SY, Jung JH, Yoon Y, Cha HJ, Lee H, Kim K, Kim J, An IS, Kim J, et al: Akt is negatively regulated by the MULAN E3 ligase. Cell Res. 22:873–885. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Suizu F, Hiramuki Y, Okumura F, Matsuda M, Okumura AJ, Hirata N, Narita M, Kohno T, Yokota J, Bohgaki M, et al: The E3 ligase TTC3 facilitates ubiquitination and degradation of phosphorylated Akt. Dev Cell. 17:800–810. 2009. View Article : Google Scholar

33 

Campbell PA and Rudnicki MA: Oct4 interaction with Hmgb2 regulates Akt signaling and pluripotency. Stem Cells. 31:1107–1120. 2013. View Article : Google Scholar : PubMed/NCBI

34 

David O, Jett J, LeBeau H, Dy G, Hughes J, Friedman M and Brody AR: Phospho-Akt overexpression in non-small cell lung cancer confers significant stage-independent survival disadvantage. Clin Cancer Res. 10:6865–6871. 2004. View Article : Google Scholar : PubMed/NCBI

35 

Lecker SH, Goldberg AL and Mitch WE: Protein degradation by the ubiquitin-proteasome pathway in normal and disease states. J Am Soc Nephrol. 17:1807–1819. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Bao B, Ahmad A, Azmi AS, Ali S and Sarkar FH: Cancer stem cells (CSCs) and mechanisms of their regulation: Implications for cancer therapy. Curr Protoc Pharmacol. 14:p.Unit-14. 252013.

37 

Gargini R, Cerliani JP, Escoll M, Antón IM and Wandosell F: Cancer stem cell-like phenotype and survival are coordinately regulated by Akt/FoxO/Bim pathway. Stem Cells. 33:646–660. 2015. View Article : Google Scholar

38 

Yu S, Shen G, Khor TO, Kim JH and Kong AN: Curcumin inhibits Akt/mammalian target of rapamycin signaling through protein phosphatase-dependent mechanism. Mol Cancer Ther. 7:2609–2620. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Jiang H, Shang X, Wu H, Gautam SC, Al-Holou S, Li C, Kuo J, Zhang L and Chopp M: Resveratrol downregulates PI3K/Akt/mTOR signaling pathways in human U251 glioma cells. J Exp Ther Oncol. 8:25–33. 2009.PubMed/NCBI

40 

Efferth T: Stem cells, cancer stem-like cells, and natural products. Planta Med. 78:935–942. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Mendoza MC, Er EE and Blenis J: The Ras-ERK and PI3K-mTOR pathways: Cross-talk and compensation. Trends Biochem Sci. 36:320–328. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Dent P: Crosstalk between ERK, AKT, and cell survival. Cancer Biol Ther. 15:245–246. 2014. View Article : Google Scholar : PubMed/NCBI

43 

McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Wong EW, Chang F, Lehmann B, Terrian DM, Milella M, Tafuri A, et al: Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochim Biophys Acta. 1773:1263–1284. 2007. View Article : Google Scholar

44 

Fresno Vara JA, Casado E, de Castro J, Cejas P, Belda-Iniesta C and González-Barón M: PI3K/Akt signalling pathway and cancer. Cancer Treat Rev. 30:193–204. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Yu CF, Liu ZX and Cantley LG: ERK negatively regulates the epidermal growth factor-mediated interaction of Gab1 and the phosphatidylinositol 3-kinase. J Biol Chem. 277:19382–19388. 2002. View Article : Google Scholar : PubMed/NCBI

46 

Bhummaphan N and Chanvorachote P: Gigantol suppresses cancer stem cell-like phenotypes in lung cancer cells. Evid Based Complement Alternat Med. 2015:8365642015. View Article : Google Scholar : PubMed/NCBI

47 

Eramo A, Lotti F, Sette G, Pilozzi E, Biffoni M, Di Virgilio A, Conticello C, Ruco L, Peschle C and De Maria R: Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ. 15:504–514. 2008. View Article : Google Scholar

48 

Sun FF, Hu YH, Xiong LP, Tu XY, Zhao JH, Chen SS, Song J and Ye XQ: Enhanced expression of stem cell markers and drug resistance in sphere-forming non-small cell lung cancer cells. Int J Clin Exp Pathol. 8:6287–6300. 2015.PubMed/NCBI

49 

Shi Y, Fu X, Hua Y, Han Y, Lu Y and Wang J: The side population in human lung cancer cell line NCI-H460 is enriched in stem-like cancer cells. PLoS One. 7:e333582012. View Article : Google Scholar : PubMed/NCBI

50 

Bertolini G, Roz L, Perego P, Tortoreto M, Fontanella E, Gatti L, Pratesi G, Fabbri A, Andriani F, Tinelli S, et al: Highly tumorigenic lung cancer CD133+ cells display stem-like features and are spared by cisplatin treatment. Proc Natl Acad Sci USA. 106:16281–16286. 2009. View Article : Google Scholar

51 

Mirza AM, Gysin S, Malek N, Nakayama K, Roberts JM and McMahon M: Cooperative regulation of the cell division cycle by the protein kinases RAF and AKT. Mol Cell Biol. 24:10868–10881. 2004. View Article : Google Scholar : PubMed/NCBI

52 

Testa JR and Tsichlis PN: AKT signaling in normal and malignant cells. Oncogene. 24:7391–7393. 2005. View Article : Google Scholar : PubMed/NCBI

53 

Dubrovska A, Kim S, Salamone RJ, Walker JR, Maira SM, García-Echeverría C, Schultz PG and Reddy VA: The role of PTEN/Akt/PI3K signaling in the maintenance and viability of prostate cancer stem-like cell populations. Proc Natl Acad Sci USA. 106:268–273. 2009. View Article : Google Scholar : PubMed/NCBI

54 

Xiang T, Ohashi A, Huang Y, Pandita TK, Ludwig T, Powell SN and Yang Q: Negative regulation of Akt activation by BRCA1. Cancer Res. 68:10040–10044. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Millimouno FM, Dong J, Yang L, Li J and Li X: Targeting apoptosis pathways in cancer and perspectives with natural compounds from mother nature. Cancer Prev Res (Phila). 7:1081–1107. 2014. View Article : Google Scholar

56 

Singh P and Bast F: Screening of multi-targeted natural compounds for receptor tyrosine kinases inhibitors and biological evaluation on cancer cell lines, in silico and in vitro. Med Oncol. 32:2332015. View Article : Google Scholar : PubMed/NCBI

57 

El Khattabi I and Sharma A: Preventing p38 MAPK-mediated MafA degradation ameliorates β-cell dysfunction under oxidative stress. Mol Endocrinol. 27:1078–1090. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Niu H, Ye BH and Dalla-Favera R: Antigen receptor signaling induces MAP kinase-mediated phosphorylation and degradation of the BCL-6 transcription factor. Genes Dev. 12:1953–1961. 1998. View Article : Google Scholar : PubMed/NCBI

59 

Deschênes-Simard X, Gaumont-Leclerc MF, Bourdeau V, Lessard F, Moiseeva O, Forest V, Igelmann S, Mallette FA, Saba-El-Leil MK, Meloche S, et al: Tumor suppressor activity of the ERK/MAPK pathway by promoting selective protein degradation. Genes Dev. 27:900–915. 2013. View Article : Google Scholar : PubMed/NCBI

60 

Ley R, Balmanno K, Hadfield K, Weston C and Cook SJ: Activation of the ERK1/2 signaling pathway promotes phosphorylation and proteasome-dependent degradation of the BH3-only protein, Bim. J Biol Chem. 278:18811–18816. 2003. View Article : Google Scholar : PubMed/NCBI

61 

Rungtabnapa P, Nimmannit U, Halim H, Rojanasakul Y and Chanvorachote P: Hydrogen peroxide inhibits non-small cell lung cancer cell anoikis through the inhibition of caveolin-1 degradation. Am J Physiol Cell Physiol. 300:C235–C245. 2011. View Article : Google Scholar :

62 

Chanvorachote P, Nimmannit U, Lu Y, Talbott S, Jiang BH and Rojanasakul Y: Nitric oxide regulates lung carcinoma cell anoikis through inhibition of ubiquitin-proteasomal degradation of caveolin-1. J Biol Chem. 284:28476–28484. 2009. View Article : Google Scholar : PubMed/NCBI

63 

Pongrakhananon V, Stueckle TA, Wang HY, O'Doherty GA, Dinu CZ, Chanvorachote P and Rojanasakul Y: Monosaccharide digitoxin derivative sensitize human non-small cell lung cancer cells to anoikis through Mcl-1 proteasomal degradation. Biochem Pharmacol. 88:23–35. 2014. View Article : Google Scholar :

64 

Pongrakhananon V, Nimmannit U, Luanpitpong S, Rojanasakul Y and Chanvorachote P: Curcumin sensitizes non-small cell lung cancer cell anoikis through reactive oxygen species-mediated Bcl-2 downregulation. Apoptosis. 15:574–585. 2010. View Article : Google Scholar : PubMed/NCBI

65 

Castor LR, Locatelli KA and Ximenes VF: Pro-oxidant activity of apocynin radical. Free Radic Biol Med. 48:1636–1643. 2010. View Article : Google Scholar : PubMed/NCBI

66 

Liu J and Mori A: Antioxidant and pro-oxidant activities of p-hydroxybenzyl alcohol and vanillin: Effects on free radicals, brain peroxidation and degradation of benzoate, deoxyribose, amino acids and DNA. Neuropharmacology. 32:659–669. 1993. View Article : Google Scholar : PubMed/NCBI

67 

Tai A, Sawano T, Yazama F and Ito H: Evaluation of antioxidant activity of vanillin by using multiple antioxidant assays. Biochim Biophys Acta. 1810:170–177. 2011. View Article : Google Scholar

Related Articles

Journal Cover

April-2017
Volume 50 Issue 4

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Srinual S, Chanvorachote P and Pongrakhananon V: Suppression of cancer stem-like phenotypes in NCI-H460 lung cancer cells by vanillin through an Akt-dependent pathway. Int J Oncol 50: 1341-1351, 2017
APA
Srinual, S., Chanvorachote, P., & Pongrakhananon, V. (2017). Suppression of cancer stem-like phenotypes in NCI-H460 lung cancer cells by vanillin through an Akt-dependent pathway. International Journal of Oncology, 50, 1341-1351. https://doi.org/10.3892/ijo.2017.3879
MLA
Srinual, S., Chanvorachote, P., Pongrakhananon, V."Suppression of cancer stem-like phenotypes in NCI-H460 lung cancer cells by vanillin through an Akt-dependent pathway". International Journal of Oncology 50.4 (2017): 1341-1351.
Chicago
Srinual, S., Chanvorachote, P., Pongrakhananon, V."Suppression of cancer stem-like phenotypes in NCI-H460 lung cancer cells by vanillin through an Akt-dependent pathway". International Journal of Oncology 50, no. 4 (2017): 1341-1351. https://doi.org/10.3892/ijo.2017.3879