![](../img/pic_top_230630.jpg?v=2501181009)
2023 Impact Factor
Glioblastoma multiforme (GBM) is the most aggressive form of adult brain tumor, classified as a grade IV astrocytoma by the World Health Organization. It constitutes approximately 40% of all primary brain tumors and 78% of malignant tumors in the central nervous system (Miller and Perry, 2007; Louis
The efficacy of radiotherapy is significantly undermined by the presence of cancer stem cells (CSCs), while the effectiveness of chemotherapy is often compromised by the overexpression of O6-methylguanine-DNA methyltransferase (MGMT), which enhances the repair of damaged DNA in GBM cells (Delello Di Filippo
Ginger (
Recent studies have highlighted that 6-gingerol enhances the sensitivity of gastric cancer cells to cisplatin, leading to cell cycle arrest, suppression of migration and invasion via the PI3K/AKT pathway, and increased radiosensitivity through G2/M arrest and apoptosis induction (Promdam and Panichayupakaranant, 2022). Additionally, it exhibits anti-proliferative effects on cervical cancer cells and induces TRAIL-mediated apoptosis in glioblastoma tumor cell lines (Lee
Despite these advances, the specific anticancer properties and mechanisms of action of 6-gingerol in GBM have not been fully elucidated. This study aims to investigate these aspects by examining the ability of 6-gingerol to cross the BBB and elucidate its mechanisms in inducing cell death in GBM cells.
Tetramethylrhodamine methyl ester (TMRM) reagent was sourced from Invitrogen (Life Technologies, CA, USA). Bovine serum albumin (BSA), RNase A, propidium iodide (PI), dihydroethidium (DHE), and the ATP Assay Kit were procured from Sigma-Aldrich (St. Louis, MO, USA). The mitogen-activated protein kinase kinase (MEK) Inhibitor U0126 was purchased from Promega (Madison, WI, USA). Mito-TEMPO and the Cell Counting Kit-8 (CCK-8) were obtained from Targetmol (Shanghai, China). The PAMPA-BBB assay kit was acquired from BioAssay Systems (Hayward, CA, USA). The Annexin V-FITC/PI apoptosis detection kit was supplied by Fremont (CA, USA). Antibodies specific to GAPDH were sourced from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Antibodies for E2F1, E2F3, p21, Cyclin A, Cyclin B, Cyclin E, Cyclin D, phospho-ERK1/2 (T202/Y204), ERK1/2, puma, Bax, Bcl-xL, Caspase-3, Caspase-9, and PARP were procured from Cell Signaling Technology (Beverly, MA, USA). Antibodies for manganese superoxide dismutase (MnSOD), RAS, RAF, phospho-RAF, EGFR, and phospho-EGFR were obtained from ABclonal (Woburn, MA, USA). Secondary horseradish peroxidase (HRP)-conjugated donkey anti-rabbit and donkey anti-mouse antibodies were purchased from Invitrogen (Life Technologies).
Human GBM cell lines M059K and U251, sourced from the American Type Culture Collection (ATCC), Manassas, VA, USA, were cultured in Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin (Gibco, Thermo Fisher Scientific, Waltham, MA, USA). The cultures were maintained at 37°C and 5% CO2 in a fully humidified environment. All experiments were conducted during the logarithmic growth phase of the cells.
Cell viability was assessed using the Cell Counting Kit-8 (CCK-8). M059K and U251 cells (4×103 cells/well) were seeded in 96-well plates overnight and then treated with various concentrations of 6-gingerol at 37°C for 24, 48, and 72 h. Post-incubation, CCK-8 solution was added, and the plates were incubated at 37°C until color development. Absorbance at 450 nm was measured using a Synergy™ HT Multi-Detection Microplate Reader (Bio-Tek Instruments, Winooski, VT, USA).
The colony formation assay followed established protocols (Tsai
The PAMPA-BBB assay is a high-throughput method for predicting a drug’s ability to penetrate the BBB. This assay evaluates the permeability of drug molecules through an artificial membrane containing porcine brain polar lipids (PBL), which mimic the composition of brain endothelial cells. The test compound diffuses from the donor well through the artificial membrane to the receptor well, and the effective permeability (Pe) is calculated based on the concentration of the compound in the receptor well. Test compounds and commercial drugs were dissolved in DMSO at a concentration of 5 mg/mL and then diluted to 25 µg/mL in PBS buffer (pH 7.4). Porcine polar brain lipid (PBL) was solubilized in dodecane at 20 mg/mL. To set up the assay, 4 µL of the PBL solution was applied to the filter surface of the donor plate. After ensuring the filter was fully saturated, 150 µL of the test compound solution was added to the donor plate, and the acceptor well was filled with 300 µL of PBS buffer. The donor plate was then carefully placed onto the acceptor plate, ensuring proper membrane and liquid contact. The assembly was covered and incubated in darkness at 25°C for 6 h. Post-incubation, the concentrations in the acceptor, donor, and reference wells were determined using a microplate reader, correlating UV absorbance with a standard curve. Effective permeability (Pe) values were then calculated, allowing compounds to be classified based on their potential to cross the BBB and indicating their possible central nervous system activity. Each sample set was tested in triplicate to ensure consistency. Compounds were classified based on their Pe values as follows: CNS+ (Pe>4.0×10–6 cm/s), CNS- (Pe<2.0×10–6 cm/s), and CNS± (Pe between 2.0×10–6 cm/s and 4.0×10–6 cm/s).
The permeability of 6-gingerol and curcumin across the BBB was evaluated using the online BBB predictor tool available at https://www.cbligand.org/BBB/index.php (Liu
For cell cycle analysis, M059K and U251 cells treated with varying concentrations of 6-gingerol for 48 h were harvested, washed with phosphate-buffered saline (PBS, pH 7.2), and fixed in 85% methanol overnight at 4°C. After centrifugation at 1500 rpm for 10 min, the cell pellet was washed twice with PBS and then resuspended in PBS containing 100 µg/mL PI and 20 Units/mL RNase A for 30 min at room temperature. A minimum of 10,000 cells per sample were analyzed using an Attune NxT flow cytometer (Thermo Fisher Scientific), with data processed using Attune NxT Flow Cytometer Software.
Apoptosis in M059K and U251 cells treated with various concentrations of 6-gingerol, alone or in combination with U0126 (10 µM), over 48 h, was assessed using the CF®488A Annexin V and PI Apoptosis Kit. Following the manufacturer’s instructions, apoptotic cells were labeled with Annexin V-FITC, and late apoptotic or necrotic cells were stained with PI. Data from 10,000 cells were acquired using a BD FACSLyric flow cytometer (BD Biosciences, San Jose, CA, USA) and analyzed with FlowJo v10 software (BD Biosciences). The analysis focused on specifically defined regions to precisely evaluate the distribution of cells across different cell cycle phases.
mtROS production and changes in ΔΨm were assessed using DHE and TMRM, respectively. After treating M059K and U251 cells with various concentrations of 6-gingerol, alone or in combination with U0126 (10 µM) or Mito-TEMPO (5 µM), for 48 h, cells were incubated with 5 µM CellROX® Green Reagent, 250 nM DHE Red Reagent, and 25 nM TMRM for 30 min at 37°C. Cells were then washed with PBS, trypsinized, and analyzed for fluorescence intensity using flow cytometry at excitation/emission wavelengths of 485/530 nm for CellROX, 510/580 nm for DHE, and 488/570 nm for TMRM. Data analysis was performed using Attune NxT Flow Cytometer Software.
Cells cultured in 10 cm dishes were treated with various concentrations of 6-gingerol for 48 h, with or without U0126 (10 µM) or Mito-TEMPO (5 µM). Proteins were extracted using RIPA buffer containing protease and phosphatase inhibitors, and total protein content was determined using a Bio-Rad Protein Assay Kit. Proteins (15-50 µg) were separated by SDS-PAGE and transferred onto PVDF membranes, which were then blocked with 5% milk in PBST, incubated with primary antibodies overnight, washed, and incubated with HRP-conjugated secondary antibodies (1:5000 dilution). Band intensities were visualized using a chemiluminescent solution (Pierce, Rockford, IL, USA) and imaged using a MultiGel-21 gel imager (Topbio, Taipei, Taiwan).
Data were analyzed using GraphPad Prism 8 software (GraphPad Software, Inc., San Diego, CA, USA). Results are presented as mean ± standard deviation (SD). Comparisons between groups were made using unpaired Student’s t-test, with a
Zingiber officinale (ginger) and Curcuma longa (turmeric), both plants of the Zingiberaceae family, contain molecular constituents such as 6-shogaol and 6-gingerol, which exhibit structural similarities to curcumin (Fig. 1A). These similarities suggest potential anticancer properties. To investigate BBB permeability, we utilized the ALzPlatform (Liu
Table 1 Predicted drug-like properties, PAMPA-BBB results (Pe: 10–6 cm/s), and predicted CNS penetration for 6-gingerol and curcumin
Compounds | Predictionb | BBB permeabilityb | |
---|---|---|---|
6-gingerol | 4.99 ± 0.53 | CNS+ | + |
Curcumin | 1.91 ± 0.03 | CNS– | – |
TMZc | 4.91 ± 0.41 | CNS+ | + |
aData are the mean ± SEM of eight independent experiments.
b[CNS+, high BBB permeation predicted; Pe (106 cm s–1)>4.0]; [CNS±, BBB permeation uncertain; Pe (106 cm s–1) from 2.0 to 4.0]; [CNS–, low BBB permeation predicted); Pe (106 cm s–1)<2.0].
cTMZ- is an oral alkylating agent used to treat GBM.
To investigate the influence of 6-gingerol on GBM cell growth, two human GBM cell lines, M059K and U251, were utilized. These cells were treated with 6-gingerol at concentrations of 25, 50, 75, 100, and 200 μM for 24, 48, and 72 h. This selection of concentrations and treatment durations was based on previously observed dose-dependent and time-dependent cytotoxic effects of 6-gingerol (Weng
Given the observed dose-dependent and time-dependent cytotoxic effects, we further investigated the impact of 6-gingerol on the cell cycle of GBM cells to understand the underlying mechanisms of its anticancer activity. Treatment with 6-gingerol resulted in an increased number of M059K and U251 cells in the G1 phase in a dose-dependent manner (Fig. 2A). This G1 arrest was associated with the accumulation of p21 and decreased levels of E2F1 (Fig. 2B). Additionally, there was a dose-dependent suppression of Cyclin D1 and Cyclin D3, with no significant changes observed in Cyclin A and Cyclin B1 levels. These findings suggest that 6-gingerol effectively induces cell cycle arrest at the G1 phase, contributing to its cytotoxic effects on GBM cells.
To determine if 6-gingerol-induced cell growth inhibition is mediated by apoptosis, we used flow cytometry with annexin V and PI staining. Annexin V detects early and late apoptosis, while PI detects late apoptosis and necrosis. Early apoptotic cells are annexin V positive and PI negative (lower right quadrant, Annexin V+/PI–), whereas late apoptotic or necrotic cells are positive for both annexin V and PI (upper right quadrant, Annexin V+/PI+). This analysis confirmed that 6-gingerol induces apoptosis in GBM cells. A significant increase in the percentage of apoptotic cells was observed in M059K and U251 cells compared to controls (Fig. 3A). This was consistent with increased activities of caspases -3 and -9 and elevated levels of cleaved PARP (Fig. 3B). There was also a decrease in Bcl-2 expression and an increase in Bax expression following exposure to 6-gingerol (Fig. 3C). Collectively, these results indicate that 6-gingerol induces apoptosis in GBM cell lines.
6-gingerol may induce ROS accumulation and ferroptosis (Liu
The MAPK pathway is pivotal in the regulation of apoptosis (Yuan
The signaling activity of the EGFR is crucial for regulating apoptosis and proliferation. Activation of EGFR leads to the autophosphorylation of specific tyrosine residues. Once activated, EGFR binds to growth factor receptor-bound protein 2 (GRB2) and recruits SH2 domain-containing transforming protein (SHC). SHC then binds to GRB2, which subsequently binds to son of sevenless homolog 1 (SOS1). SOS1 activates RAS, which in turn activates RAF-1. RAF-1 phosphorylates MEK 1/2, leading to the activation of ERK 1/2, culminating in various biological responses. EGFR plays a pivotal role in coordinating apoptosis and proliferation by mediating the cell-to-cell propagation of ERK activation.
The effect of 6-gingerol on EGFR expression and phosphorylation was investigated. Examination of the upstream regulators of ERK, specifically the EGFR-RAS-RAF axis, showed that 6-gingerol treatment significantly decreased phosphorylated EGFR, RAS levels, and phosphorylated RAF (Fig. 6A). This suggests that 6-gingerol exerts its cytotoxic effects through the RAF-1/MEK/ERK pathway.
The role of the EGFR agonist in this signaling pathway was further investigated by analyzing the expression of these signaling molecules in the presence of EGF and 6-gingerol. Increased p-EGFR and p-ERK1/2 expression was noted upon EGF treatment, observed after 30 min, suggesting the role of an EGFR agonist in the activation of this signaling pathway. However, 6-gingerol reversed the effect of EGF, indicating that the antiproliferative and anti-apoptotic activities of 6-gingerol against GBM cells involve modulation of EGFR signaling (Fig. 6B).
Brain cancer, particularly GBM, is the most prevalent malignancy within the central nervous system and ranks among the top causes of cancer-related deaths globally (Miller and Perry, 2007; Louis
Combretastatin A-4 (CA-4), for instance, induces G2 arrest in U-87 cells by increasing cellular ROS levels (Roshan
Despite extensive research on curcumin, 6-gingerol has shown similar efficacy in inhibiting lung cancer cells, suggesting its potential as an anti-cancer agent (Eren and Betul, 2016). Moreover, 6-gingerol uniquely induces autophagy-dependent ferroptosis by inhibiting the expression of ubiquitin-specific peptidase 14 (USP14) (Tsai
Recent studies have unveiled the potential of 6-gingerol in sensitizing gastric and epithelial ovarian cancer cells to cisplatin, reducing the proliferation of cervical cancer cells, and inducing TRAIL-mediated apoptosis in glioblastoma cell lines (Czarnik-Kwaśniak
Traditionally, MnSOD is viewed as a tumor suppressor (Zhong
The RAS/RAF/MEK/ERK signaling pathway is crucial in cancer development and progression, involving the G-protein RAS and kinases RAF, MEK, and ERK (Song
Our study also highlights 6-gingerol ability to cross the BBB and exert potent cytotoxic effects on GBM cells. It induces G1 cell cycle arrest and apoptosis by modulating key cell cycle regulators and increasing pro-apoptotic signals. Specifically, 6-gingerol enhances G1 phase arrest through upregulation of p21 and downregulation of E2F1, along with suppression of Cyclin D1 and Cyclin D3. It also triggers apoptosis by increasing caspase-3 and -9 activities and altering the balance of Bcl-2 and Bax. Furthermore, 6-gingerol impacts mitochondrial function by inducing mtROS and disrupting ΔΨm, promoting oxidative stress-mediated apoptosis (Fig. 7).
Significantly, 6-gingerol inhibition of the EGFR/RAS/RAF/ERK signaling pathway underscores its mechanism of action, presenting a comprehensive approach to suppressing GBM cell proliferation and survival. These results underscore the potential of 6-gingerol as a multifaceted therapeutic candidate for GBM, warranting further investigation into its clinical applications.
This research was supported by grants MOST-111-2314-B-037-095-MY2 (C.-Y.T.). The present research was supported by a grant from the Chi-Mei Medical Center and Kaohsiung Medical University Research Foundation (112CM-KMU-08) (C.-Y.T.), grants KMUH111-1T02 from Kaohsiung Medical University hospital (C.-Y.T.) and grants CCFHR11302 from Chi-Mei Medical Center (S.-W.L.).
The authors assert that there are no known financial conflicts of interest or personal relationships that could be perceived as affecting the integrity of the work presented in this paper.
Sher-Wei Lim: Methodology, Conceptualization, Writing – review & editing. Wei-Chung Chen: Methodology, Investigation, Formal analysis, Writing – review & editing. Huey-Jiun Ko: Methodology, Writing – review & editing, Validation, Data curation. Yu-Feng Su: Visualization, Supervision, Methodology. Chieh-Hsin Wu: Project administration, Writing – review & editing. Fu-Long Huang: Data curation, Writing – review & editing. Chien-Feng Li: Project administration, Resources. Cheng Yu Tsai: Project administration, Methodology, Resources, Writing – review & editing.
![]() |
![]() |