Research Article

Pyrogallol induces G0/G1 cell cycle arrest and apoptosis through mitochondrial pathway in PC-3 human prostate cancer cells

Seong-Min Hong1https://orcid.org/0000-0002-7646-3138, Hyun-Dong Cho2https://orcid.org/0000-0001-8756-3217, Kwon-Il Seo3,*https://orcid.org/0000-0002-2391-9146
Author Information & Copyright
1College of Pharmacy, Gachon University, Incheon 21936, Korea
2Department of Food and Nutrition, Sunchon National University, Suncheon 57922, Korea
3Department of Food Biotechnology, Dong-A University, Busan 49315, Korea
*Corresponding author Kwon-Il Seo, Tel: +82-51-200-7565, E-mail: kseo@dau.ac.kr

Citation: Hong SM, Cho HD, Seo KI. Pyrogallol induces G0/G1 cell cycle arrest and apoptosis through mitochondrial pathway in PC-3 human prostate cancer cells. Food Sci. Preserv., 32(3), 486-497 (2025)

Copyright © The Korean Society of Food Preservation. This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Received: Feb 13, 2025; Revised: Mar 25, 2025; Accepted: Mar 28, 2025

Published Online: Jun 30, 2025

Abstract

Pyrogallol (PG), a naturally occurring dihydroxyphenol, was shown to suppress the proliferation of numerous cancer cell lines (e.g., lymphoma, lung, and breast cancer cells). However, its effects on human prostate cancer remain largely unexplored. Thus, we evaluated the anti-cancer effects of PG in prostate cancer cells (i.e., PC-3, RC-58T, and LNCaP) and a xenograft mouse model. In the in vitro assays, cells were treated with PG at various concentrations (2.5-200 μM) for 1 day, and the half-maximal inhibitory concentration (IC50) values were determined to be 55.53 μM for PC-3, 65.03 μM for RC-58T, 71.78 μM for LNCaP, and >200 μM for RWPE-1. Mechanistic analyses on PC-3 cell lines revealed that PG induced mitochondrial-dependent apoptosis, as evidenced by increased expression of apoptosis signaling (i.e., Bax, cytochrome C, caspase-3, and PARP), and decreased expression of anti-apoptosis signaling as Bcl-2. Additionally, PG modulated MAPK pathways, which were associated with the activation of caspase-3 and caspase-8. Furthermore, PG treatment induced G0/G1 phase cell cycle arrest. In our in vivo model, groups treated with PG (2.5-5 mg/kg, p.o.) for 2 weeks exhibited a significant reduction in tumor growth, along with increased expression of apoptotic signaling markers in tumor tissues. We suggest that PG may serve as a promising therapeutic agents for prostate cancer, highlighting their potential applications in phytomedicine and phytotherapy.

Keywords: apoptosis; cell cycle arrest; prostate cancer; pyrogallol

1. Introduction

Prostate cancer is the diagnosed malignancy in man in the world, and remains a major risk of cancer-related mortality, following lung and colorectal cancer (Siegel et al., 2024). In the past decade, more than two million men in the United States have been diagnosed with this disease, with incidence rates have also rising in Asian populations (Panaiyadiyan and Kumar, 2024; Siegel et al., 2024). Current therapeutic options, including androgen deprivation, hormone therapy, immunotherapy, radiopharmaceuticals, and cytotoxic agents, have shown significant efficacy (Guinney et al., 2017; Sonpavde and Kantoff, 2012; Tanimoto et al., 2010). However, these treatments often lead to adverse effects, including damage to healthy tissues and the development of drug resistance (Frederiks et al., 2015; Karavelioglu et al., 2016), ultimately allowing unresponsive tumors to progress to a metastatic stage. Increasing evidence suggests that natural bioactive compounds, including plant-derived extracts and phytochemicals, offer promising alternatives due to their lower toxicity and potential biocompatibility in body (Kelloff et al., 2000; Lozano-Casabianca et al., 2022). As a result, there is growing interest in studying dietary natural products with cancer-preventive properties, as elucidating their molecular mechanisms could aid in the development of more effective cancer treatments.

Pyrogallol (PG), a naturally occurring catechin compound (benzene-1,2,3-triol), is a polyphenol widely found in plants such as sorghum (Djanaguiraman et al., 2018), green tea (Liu et al., 2022), and gooseberry (Ptak et al., 1986). In recent years, several studies have demonstrated the biological properties of PG, with a particular focus on its cytotoxic effects against cancer cells. It has been shown to induce to cell death in various malignancies, including lymphoma (Saeki et al., 2000), human lung cancer cells (Han et al., 2009a; Han et al., 2009b), human gastric cells (Park et al., 2008), and human breast cancer cells (Nemec et al., 2016). Beyond its anticancer properties, PG has demonstrated antimicrobial activity (Lima et al., 2016), anti-inflammatory effects in bronchial epithelial cells (Nicolis et al., 2008), and inhibition of vascular smooth muscle cell migration in carotid ligation models (Ma et al., 2016). Although PG has shown promising effects, the specific molecular pathways underlying its impact on human prostate cancer cells are not yet well understood.

Cell cycle regulation is crucial for cancer progression, enabling tumor cells to transition from a quiescent (G0) state to active proliferation and metastasis while maintaining genomic stability (Kim, 2021; Rahman et al., 2021). Notably, abnormalities in cyclin-dependent kinase (CDK) regulation across different phases in cell cycle led to increased efforts to identify small-molecule inhibitors that specifically target these kinases. Five key CDKs govern cell cycle progression: CDK4, CDK6, and CDK2 in G1; CDK2 in S; and CDK1 in G2 and M phases. Each phase requires specific cyclins to complex with CDKs, facilitating precise cell cycle transitions (Chen et al., 2017; Roh et al., 2016; Wang et al., 2017). At the same time, apoptosis, which is a key progress in the cell death, plays a vital role in removing damaged or abnormal cells from the body. Apoptotic events are characterized by several factors (e.g., DNA fragmentation, chromatin condensation, and increasing the apoptotic bodies (Walker et al., 2016). Links between apoptosis and cell cycle regulation have been established, with tumor suppressor proteins such as p53 acting as key regulators (Pucci et al., 2000). Since several proteins involved in cell death also regulate proliferative signaling, compounds that can induce the cell cycle arrests and trigger apoptosis to apoptosis hold great promise as potential cancer therapeutics.

This study examines the potential effects of PG in prostate cancer cells derived from human (PC-3) and a C57BL/6 nude mouse xenograft model, focusing on its underlying mechanisms. In particular, we investigated how PG induces the apoptosis signaling and cell cycle arrest for assessing its potential as a prostate cancer therapeutic agent. Gaining a deeper understanding of the molecular mechanisms behind PG’s effects could contribute to the development of new prostate cancer treatments derived from natural products.

2. Materials and methods

2.1. Chemicals

Dulbecco’s Modified Eagle Medium (DMEM), fetal bovine serum (FBS), and penicillin-streptomycin, were obtained from Gibco BRL Co. (Gaithersburg, MD, USA). The pan-caspase inhibitor (z-VAD-fmk) was sourced from R&D Systems (Minneapolis, MN, USA). Antibodies targeting cyclin-dependent kinase 2 (CDK2), CDK4, Cyclin D1, p53, p21, Bax, Bcl-2, cytochrome C, caspase-3, PARP, and α-tubulin were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Extracellular-signal-regulated kinase (ERK), p-ERK, c-Jun N-terminal kinase (JNK), p-JNK, p38, p-p38, and caspase-8 were purchased from Cell Signaling Technologies (Beverly, MA, USA). Chemiluminescence detection kit for analyzing western blotting assay was obtained from Amersham Life Science (Amersham, Buckinghamshire, UK). Pyrogallol (PG) and other chemicals were purchased from Sigma-Aldrich Co. Ltd., St. Louis, MO, USA For all experiments, PG was dissolved in distilled water at a concentration of 100 mM for storage at 4°C before use in experimental procedures.

2.2. Prostate cancer and epithelial cell culture

All cancer cell lines derived from human [i.e., LNCaP-FGC (lymph node metastasis), PC-3 (bone metastasis), and RWPE-1 (normal epithelial cells)], were acquired at American Type Culture Collection (ATCC, Rockville, MD, USA). In addition, primary cell (RC-58T/h/SA#4) was obtained from the Center for Prostate Disease Research (CPDR). LNCaP, PC-3, and RC-58T/h/SA#4 cells were cultured in DMEM with 10% FBS and 1% penicillin-streptomycin (Gibco, USA). RWPE-1 cells were maintained in Keratinocyte-SFM with 10% FBS and 1% penicillin-streptomycin. Each cell was incubated at 37°C in 5% CO2.

2.3. Measurement of cell viability

This experiment was assessed via sulforhodamine B (SRB) assay (Sigma-Aldrich Co.), according to previous study (Kim et al., 2016). Each cell was seeded (2.5×104 cells/well) in 48-well plates and treated with PG at different concentrations for 24 h. After treatment, the cells fixed by treating 10% trichloroacetic acid at 4°C for 1 h. Plates were then stained with 0.4% (w/v) SRB reagent. The stained cells were removed using 1% acetic acid and then dissolved using Tris (10 mM). The dissolved solution was detected at 540 nm with a microplate reader (Molecular Devices, Inc., San Jose, CA, USA). IC50 values for PG were represented using GraphPad Prism 5 software (GraphPad Software Co., San Diego, CA, USA). For investigating the role of caspase inhibition, we used z-VAD-fmk as a caspase inhibitor.

2.4. Analysis of cell cycle and apoptotic cell population by flow cytometry

Cell cycle progression and apoptosis were analyzed as following the protocol provided by Merck Millipore Co. (Burlington, MA, USA). PC-3 cells were seeded into six-well plates at the recommended density and treated with PG for 24 h. For cell cycle analysis, cells were detached using trypsin-EDTA and fixed at −20°C for 3 h. Following fixation, a staining reagent (200 μL) was added to the cells and incubated for 30 min at room temperature. To assess apoptosis, cells stained by using Annexin V staining solution and incubated during 30 min at room temperature. Flow cytometry was performed on 5,000 gated events per sample, and cell cycle distribution was analyzed based on fluorescence intensity.

2.5. Detection of chromatin condensation

The changes in nuclear fragmentation and chromatin condensation were observed using bis-Benzimide (Hoechst 33258) chemicals under fluorescence microscopy. Cells were seeded (1×105 cells/mL) in six-well plates and exposed to PG for 24 h. After treatment, cells were harvested and reacted with bis-Benzimide (5 μg/mL) during 20 min at room temperature. An aliquot of the cell suspension was placed on a glass slide and observed via fluorescence microscope (Olympus Optical Co. Ltd., Japan)

2.6. Western blotting assay

For the apoptosis pathway investigation, cells were seeded in 100 mm dishes and treated with various concentrations of PG for 24 h. In contrast, to assess the activation of mitogen-activated protein kinase (MAPK) pathways, cells were similarly seeded in 100 mm dishes and exposed to PG at the same concentration range for 30 min. Cells or tumor tissues collected from mice were lysed by using lysis buffer (49 mM Tris-HCl, 149 mM NaCl, 1 mM EDTA, 49 mM NaF, 29 mM Na4P2O7, 1 mM PMSF, and 2 μg/mL aprotinin) during 1 h at 4°C. The protein concentration was calculated via bicinchoninic acid (BCA) protein assay kit (Pierce Biotechnology Co., Waltham, MA, USA). Equal amounts of protein (5 μg) were separated on 10-12% SDS-PAGE gels and transferred onto nitrocellulose membranes. The membranes were blocked with 5% bovine serum albumin (BSA) and then incubated with primary antibodies (1:1000) overnight at 4°C. In the next day, each membrane was washed and incubated with secondary antibodies (1:2000) during at 4°C. The bands of protein were detected using an ECL kit (Santa Cruz, CA, USA), and band intensities were measured with Image StudioTM Lite software (LI-COR Inc., NE, USA), with normalization to α-tubulin levels.

2.7. Human prostate tumor xenograft mouse model

All animal procedures were conducted following the guidelines approved by Dong-A University’s Institutional Animal Care and Use Committee (DIACUC-17-15, approved in Sep. 2017). Male C57BL/6 athymic mice (five weeks old) were maintained under pathogen-free conditions. Mice were fed ad libitum access to an AIN-93G diet and sterilized water. Each mouse (Eight mice per each group) randomly divided as follows: (1) Control group by treating distilled water (p.o.); (2) 2.5 mg/kg/day of PG-treated group (p.o.); (3) 5 mg/kg/day PG treated group (p.o.). PG administration started on day 6, once tumors reached 20-25 mm2 volumes. The treatments of PG were freshly prepared daily and administered via oral gavage (p.o.) for 2 weeks. Tumor volume was measured daily using calipers and calculated as follows: length/2 × width2. Also, collected tumor from mice were measured the expression of protein levels in Bax, Bcl-2, PARP, and Caspase-3.

2.8. Statistical analysis

All data are represented as the mean±standard error (SE) based on a minimum of three independent experiments (n=3-8). Statistical analysis was performed using one-way analysis of variance (ANOVA), followed by Duncan’s multiple range test for post-hoc comparisons. Differences were considered statistically significant at *p<0.05, **p<0.01, and ***p<0.001.

3. Results and discussion

3.1. PG affects human prostate cancer cells in comparison with human prostate epithelial cells

Pyrogallol (PG; C6H6O3) is a naturally occurring phenolic compound present in various plants, such as sorghum, green tea, gooseberries, and hardwood species. Previous studies have demonstrated PG’s ability to inhibit the proliferation of several cancer cell types (Arjsri et al., 2023; Han et al., 2009a; Park et al., 2008; Saeki et al., 2000); however, its precise mechanisms in prostate cancer remain unclear. Many plant-derived compounds have been explored for their potential in cancer treatment due to their lower toxicity in normal cells and ability to selectively target malignant cells (Lee et al., 2012; Naeem et al., 2022). Therefore, we assessed the anti-cancer activity of PG in prostate cancer cells and investigated its underlying mechanisms.

To determine the effect of PG on prostate cancer cell proliferation, SRB assays were performed after treating cells with PG at varying concentrations (2.5-200 μM) for 24 h (Fig. 1). As illustrated in Fig. 1A and 1B, PG treatment significantly reduced cell viability in a dose-dependent manner in prostate cancer cells, whereas RWPE-1 prostate epithelial cells exhibited no significant decline in proliferation. The IC50 values of PG were calculated as 55.53 μM for PC-3, 65.03 μM for RC-58T, and 71.78 μM for LNCaP, while RWPE-1 cells had an IC50 exceeding 200 μM (Fig. 1C). Interestingly, our results showed that PG exerted prostate cancer-specific cytotoxic effects in PC-3 cells, a castration-resistant prostate cancer cell line, when compared to normal prostate epithelial cells (RWPE-1). These findings suggest that PG preferentially inhibits the proliferation of prostate cancer cells while exhibiting minimal cytotoxicity toward normal epithelial cells, indicating its selective anti-cancer potential.

kjfp-32-3-486-g1
Fig. 1. Cell growth inhibitory effects in prostate cancer and prostate epithelial cells treated with PG for 24 h. (A), Cell viability was measured by SRB assay. All values are mean±SE (n=3). (B), After 24 h incubation with PG, cell morphology was visualized by inverted microscopy (×200). (C), IC50, values of cells were identified by Graph Prism 5. *p<0.05, **p<0.01, and ***p<0.001 vs. Control group (One-way ANOVA).
Download Original Figure
3.2. PG induces apoptosis and G0/G1 arrest with modulation of cell cycle-related protein expression in PC-3 cells

Apoptosis involves cellular changes such as chromatin condensation, apoptotic body formation, DNA fragmentation, and protein degradation (Elmore, 2007). Both apoptosis induction and cell cycle arrest are considered essential mechanisms in cancer treatment, as they contribute to inhibiting uncontrolled cell proliferation (Lin et al., 2013; Naeem et al., 2022; Preya et al., 2017; Semprebon et al., 2015). To investigate whether the anti-proliferative effects of PG were associated with apoptosis, annexin V staining, DNA fragmentation analysis, and Hoechst staining were performed. As shown in Fig. 2A, treatment with PG for 24 h significantly increased the late apoptotic cell population in a dose-dependent manner. Cells undergoing apoptosis exhibited characteristic morphological changes, including membrane blebbing, cytoplasmic condensation, nuclear fragmentation, and chromatin condensation (Erekat, 2022). Additionally, DNA fragmentation and chromatin condensation were detected in PC-3 cells following treatment with 50-100 μM PG for 24 h (Fig. 2B and 2C), providing further evidence of apoptosis induced by PG.

kjfp-32-3-486-g2
Fig. 2. PG induces apoptotic cell death in PC-3 prostate cancer cells. Cells were treated with PG for 24 h. (A), Early and late apoptosis population was analyzed by Annexin V staining assay. All values are mean±SE (n=3). (B), DNA fragmentation was observed by 2% agarose gel electrophoresis. (C), Nuclear condensation in response to PG treatment as detected by Hoechst staining assay. *p<0.05 and **p<0.01 vs. Control group (One-way ANOVA).
Download Original Figure

Cell cycle regulation plays a crucial role in maintaining cellular homeostasis, and disruptions in this process contribute to cancer progression (Vermeulen et al., 2003). CDKs, cyclins, p21, and p53 are key regulators of cell cycle checkpoints at G1/S and G2/M phases, and inhibiting these transitions can prevent cancer cell proliferation (Kastan and Bartek, 2004). PG has previously been reported to induce cell cycle arrest in lung and cervical cancer cells (Han et al., 2009b; Kim et al., 2007; Yang et al., 2009). To examine the impact of PG on cell cycle progression, flow cytometry analysis was conducted following 24 h of treatment. As shown in Fig. 3, PG treatment led to a dose-dependent accumulation of PC-3 cells in the G0/G1 phase, accompanied by a corresponding decrease in the proportion of cells in the S and G2/M phases. These results indicate that PG inhibits prostate cancer cell proliferation by inducing G0/G1 cell cycle arrest.

kjfp-32-3-486-g3
Fig. 3. PG induces G0/G1 cell cycle arrest in PC-3 prostate cancer cells. Cells were treated with PG for 24 h. (A), Cell cycle population was analyzed by flow cytometry. (B), Expression of cell cycle arrest proteins in PC-3 cells. All values are mean±SE (n=3). *p<0.05 and **p<0.01 vs. Control group (One-way ANOVA).
Download Original Figure
3.3. PG induced apoptosis and modulated MAPKs pathways in PC-3 cells via caspase-dependent and inhibited tumor growth in a xenograft prostate cancer model

The Bcl-2 family of proteins regulates mitochondrial apoptosis by modulating cytochrome C release and PARP cleavage in response to cellular stress (Chaitanya and Babu, 2009). The caspase cascade, which includes intrinsic and extrinsic pathways, plays a pivotal role in apoptosis regulation (Hengartner, 2000). Previous research has linked PG-induced apoptosis in epithelial cells to caspase-3 activation (He et al., 2012) and demonstrated its apoptotic effects in lung cancer cells through caspase and PARP cleavage (Han et al., 2009a).

To examine the apoptotic mechanism of PG, the expression of key apoptosis-related proteins was analyzed. In PC-3 cells, PG treatment resulted in an upregulation of the pro-apoptotic protein Bax and a downregulation of the anti-apoptotic protein Bcl-2. Additionally, PG-treated cells exhibited higher levels of cytochrome C and cleaved-PARP, while cleaved caspase-3 expression was significantly reduced compared to untreated controls. Moreover, as shown in Fig. 4B, pre-treatment with the pan-caspase inhibitor z-VAD-fmk completely abolished PG-induced apoptosis in a dose-dependent manner. These findings confirm that PG-induced apoptosis in PC-3 cells is caspase-dependent.

kjfp-32-3-486-g4
Fig. 4. PG induces caspase-dependent apoptosis in PC-3 cells by activating the mitochondrial apoptotic pathway. (A), Expression of Bax, Bcl-2, cytochrome C, PARP, and caspases-3 proteins in PC-3 cells. Cells were treated with PG for 24 h. Total cell lysates were subjected to western blotting to detect expression levels of proteins. (B), Effects of the caspase inhibitors z-vad-fmk on PG-induced cell death in PC-3 cells. Cells were pretreated with 10 μM z-vad-fmk for 2 h and then incubated with 25, 50, and 100 μM PG for 24 h. All values are mean±SE (n=3). *p<0.05, **p<0.01, and ***p<0.001 vs. Control group (One-way ANOVA).
Download Original Figure

MAPKs pathways, including p-ERK, p38, and JNK, regulate key cellular functions such as survival, apoptosis, and differentiation (Rodríguez-Berriguete et al., 2012). Generally, elevated JNK and p38 signaling are linked with cell death and tumor suppression, whereas increased ERK signaling promotes cell survival and tumor progression (Rodríguez-Berriguete et al., 2010; Yap et al., 2008). To further investigate the effects of PG, we evaluated its impact on MAPK phosphorylation after a 30-min exposure (Fig. 5A). PG treatment resulted in a concentration-dependent increase in JNK expression, while phosphorylated ERK and p38 levels were significantly reduced. Previous studies have demonstrated that the JNK cascade is pivotal in apoptosis through the regulation of Bcl-2 family proteins, with several anti-prostate cancer agents downregulating Bcl-2 via JNK signaling in PC-3 cells. Our findings suggest that PG promotes the degradation of Bcl-2 and activates caspase-3 through JNK pathway regulation, which leads to cell cycle arrest. Additionally, the expressed levels of p38 significantly decreased by treating PG. Elevated p38 activation is associated with pro-apoptotic effects for survival signal, implied that PG may not be primarily mediated through p38 signaling.

kjfp-32-3-486-g5
Fig. 5. PG modulates MAPK signaling, including the JNK and p38 pathways, and influences extrinsic apoptosis via caspase-8 activation. (A), Expression of p-ERK/ERK, p-p38/p38, p-JNK/JNK proteins in PC-3 cells. Cells were treated with PG for 30 min. Total cell lysates were subjected to western blotting to detect expression levels of proteins. (B), Expression of caspase-8 proteins in PC-3 cells. Cells were treated with PG for 24 h. Total cell lysates were subjected to western blotting to detect expression levels of proteins. Data are mean±SE (n=3). *p<0.05, **p<0.01, and ***p<0.001 vs. Control group (One-way ANOVA).
Download Original Figure

Elevated ERK signaling is frequently observed in various tumors, including prostate cancer, and is associated with apoptosis pathway (Bos, 1989). Previous studies has demonstrated that phosphorylated ERK levels associate with tumor grade and relapse in primary prostate cancer (Jull et al., 2001). Furthermore, phytochemicals such as formononetin have been shown to inhibit PC-3 cell proliferation by inactivating ERK signaling, which leads to increased expression of Bax protein (Ye et al., 2012). Our findings suggest that PG may exert its anti-cancer effects on human prostate cancer cells through modulation of the ERK-Bax pathway. In addition, PG treatment significantly elevated the expression levels of caspase-8 as a key extrinsic apoptotic marker critical for initiating programmed cell death. The activation of JNK signaling is linked to the regulation of capase-8-mediated apoptosis (Xu and Hu, 2020). As shown in Fig. 5B, the treatment of PG at various concentrations significantly increased caspase-8 expression, suggesting that PG may be play a potential role in modulating the JNK-Caspase-8 pathways.

To further examine PG’s anti-tumor potential, its effects were tested in a xenograft prostate cancer model using athymic mice. PC-3 xenografts were established, and mice were administered PG (2.5-5 mg/kg/day) via oral gavage for 14 days. PG treatment significantly reduced tumor weight and volume without causing significant changes in body weight (Fig. 6A). As illustrated in Fig. 6B, control mice had an average tumor weight of 3.13±1.24 g, whereas PG-treated mice exhibited a significantly lower average tumor weight of 0.49±0.4 g. Similarly, PG-treated mice showed a substantial reduction in tumor volume compared to the control group (Fig. 6C). Furthermore, we assessed the expression levels of apoptosis markers including Bax, Bcl-2, cleaved PARP, and caspase-3 in tumor tissues. As shown in Fig. 6D, the PG-treated groups exhibited expression levels of Bax, Bcl-2, cleaved PARP, and caspase-3 that were consistent with our in vitro findings. These results support the potential of PG as an effective anti-prostate cancer candidate.

kjfp-32-3-486-g6
Fig. 6. PG suppresses the growth of PC-3 human prostate cancer cells in a xenograft athymic mouse model. C57BL/6 athymic mice were subcutaneously inoculated with PC-3 cells. Administration of vehicle and PG (2.5 and 5 mg/kg/day) was initiated after tumor volume reached 15-20 mm3. (A), Tumors were excised from mice 22 days after cell inoculation and photographed. (B), Tumor weights are represented as the mean±SE of eight mice. (C), Tumor volumes were measured in two dimensions and calculated as follows: length/2 × width2. (D), Expression of Bax, Bcl-2, cytochrome C, PARP, and caspases-3 proteins in tumor tissues from mice. Total tumor tissues lysates were subjected to western blotting to detect expression levels of proteins. Tumor volumes are mean±SE (n=8). *p<0.05 and **p<0.01 vs. Control group (One-way ANOVA).
Download Original Figure

Previous studies have reported an oral LD50 for PG of approximately 800-1,270 mg/kg (Berndt et al., 1991). Consequently, we anticipated that daily administration of PG at doses of 2.5 and 5 mg/kg would effectively suppress prostate tumor growth in vivo without inducing any toxic effects. Although numerous therapeutic agents (e.g., abiraterone acetate, enzalutamide, apalutamide, etc.) are currently employed in the management of prostate cancer (Crawford et al., 2018), the potential for synergistic interactions between these drugs and pyrogallol has not been previously explored. Thus, we propose that future study should be investigated whether combining PG with established prostate cancer therapies may enhance anti-prostate cancer effect via synergistic mechanisms.

4. Conclusions

This study offers new insights into the anti-cancer effects of PG in prostate cancer, both in vitro and in vivo. Our results indicate that PG induces apoptosis in PC-3 prostate cancer cells through mitochondrial-mediated pathways while also causing cell cycle arrest at the G0/G1 phase. Additionally, PG treatment exhibits caspase-3 activation and MAPKs signaling, reinforcing its role in apoptotic regulation. In a xenograft mouse model, PG administration significantly reduced tumor growth and increased apoptosis markers such as Bax/Bcl-2, cleaved PARP, and caspase-3, suggesting its potential as a therapeutic agent. These findings highlight PG as a promising candidate for the treatment and prevention of prostate cancer. Further studies are required to evaluate its clinical applications and its potential synergy with existing therapies.

Acknowledgements

None.

Conflict of interests

The authors declare no potential conflicts of interest.

Author contributions

Conceptualization: Seo KI. Methodology: Hong SM, Cho HD. Formal analysis: Hong SM. Validation: Hong SM. Writing - original draft: Hong SM. Writing - review & editing: Hong SM, Seok KI.

Ethics approval

This research was approved by IACUC from the Dong-A University (approval no. DIACUC-17-15, approved in Sep. 2017).

Funding

None.

ORCID

Seong-Min Hong (First author) https://orcid.org/0000-0002-7646-3138

Hyun-Dong Cho https://orcid.org/0000-0001-8756-3217

Kwon-Il Seo (Corresponding author) https://orcid.org/0000-0002-2391-9146

References

1.

Arjsri P, Mapoung S, Semmarath W, Srisawad K, Tuntiwechapikul W, Yodkeeree S, Dejkriengkraikul P. Pyrogallol from spirogyra neglecta inhibits proliferation and promotes apoptosis in castration-resistant prostate cancer cells via modulating akt/gsk-3beta/beta-catenin signaling pathway. Int J Mol Sci. 24:6452 2023;

2.

Berndt WO, Bergfeld WF, Boutwell RK, Carlton WW, Hoffmann DK, Schroeter AL, Shank RC. Final report on the safety assessment of pyrogallol. J American Coll Toxicol. 10:67-85 1991;

3.

Bos JL. Ras oncogenes in human cancer: A review. Cancer Res. 49:4682-4689 1989;

4.

Chaitanya GV, Babu PP. Differential parp cleavage: An indication of heterogeneous forms of cell death and involvement of multiple proteases in the infarct of focal cerebral ischemia in rat. Cell Mol Neurobiol. 29:563-573 2009;

5.

Chen XX, Leung GPH, Zhang ZJ, Xiao JB, Lao LX, Feng F, Mak JCW, Wang Y, Sze SCW, Zhang KYB. Proanthocyanidins from uncaria rhynchophylla induced apoptosis in mda-mb-231 breast cancer cells while enhancing cytotoxic effects of 5-fluorouracil. Food Chem Toxicol. 107:248-260 2017;

6.

Crawford ED, Schellhammer PF, Mcleod DG, Moul JW, Higano CS, Shore N, Denis L, Iversen P, Eisenberger MA, Labrie F. Androgen receptor targeted treatments of prostate cancer: 35 years of progress with antiandrogens. J Urol. 200:956-966 2018;

7.

Djanaguiraman M, Perumal R, Jagadish SVK, Ciampitti IA, Welti R, Prasad PVV. Sensitivity of sorghum pollen and pistil to high-temperature stress. Plant Cell Environ. 41:1065-1082 2018;

8.

Elmore S. Apoptosis: A review of programmed cell death. Toxicol Pathol. 35:495-516 2007;

9.

Erekat NS. Programmed cell death in diabetic nephropathy: A review of apoptosis, autophagy, and necroptosis. Med Sci Monit. 28:e937766 2022;

10.

Frederiks CN, Lam SW, Guchelaar HJ, Boven E. Genetic polymorphisms and paclitaxel- or docetaxel-induced toxicities: A systematic review. Cancer Treat Rev. 41:935-950 2015;

11.

Guinney J, Wang T, Laajala TD, Winner KK, Bare JC, Neto EC, Khan SA, Peddinti G, Airola A, Pahikkala T, Mirtti T, Yu T, Bot BM, Shen L, Abdallah K, Norman T, Friend S, Stolovitzky G, Soule H, Sweeney CJ, Ryan CJ, Scher HI, Sartor O, Xie Y, Aittokallio T, Zhou FL, Costello JC. Prostate Cancer Challenge DC. Prediction of overall survival for patients with metastatic castration-resistant prostate cancer: Development of a prognostic model through a crowdsourced challenge with open clinical trial data. Lancet Oncol. 18:132-142 2017;

12.

Han YH, Kim SH, Kim SZ, Park WH. Pyrogallol inhibits the growth of human pulmonary adenocarcinoma a549 cells by arresting cell cycle and triggering apoptosis. J Biochem Mol Toxicol. 23:36-42 2009a;

13.

Han YH, Kim SZ, Kim SH, Park WH. Pyrogallol inhibits the growth of lung cancer calu-6 cells via caspase-dependent apoptosis. Chem Biol Interact. 177:107-114 2009b;

14.

He D, Ma X, Chen Y, Cai Y, Ru X, Bruce IC, Xia Q, Shi G, Jin J. Luteolin inhibits pyrogallol-induced apoptosis through the extracellular signal-regulated kinase signaling pathway. FEBS J. 279:1834-1843 2012;

15.

Hengartner MO. The biochemistry of apoptosis. Nature. 407:770-776 2000;

16.

Jull B, Plummer H, Schuller H. Nicotinic receptor-mediated activation by the tobacco-specific nitrosamine nnk of a raf-1/map kinase pathway, resulting in phosphorylation of c-myc in human small cell lung carcinoma cells and pulmonary neuroendocrine cells. J Cancer Res Clin Oncol. 127:707-717 2001;

17.

Karavelioglu E, Gonul Y, Aksit H, Boyaci MG, Karademir M, Simsek N, Guven M, Atalay T, Rakip U. Cabazitaxel causes a dose-dependent central nervous system toxicity in rats. J Neurol Sci. 360:66-71 2016;

18.

Kastan MB, Bartek J. Cell-cycle checkpoints and cancer. Nature. 432:316-323 2004;

19.

Kelloff GJ, Crowell JA, Steele VE, Lubet RA, Malone WA, Boone CW, Kopelovich L, Hawk ET, Lieberman R, Lawrence JA, Ali I, Viner JL, Sigman CC. Progress in cancer chemoprevention: Development of diet-derived chemopreventive agents. J Nutr. 130:467S-471S 2000;

20.

Kim GD. Harmine hydrochloride triggers g2/m cell cycle arrest and apoptosis in hct116 cells through erk and pi3k/akt/mtor signaling pathways. Prev Nutr Food Sci. 26:445-452 2021;

21.

Kim NH, Kim K, Park WS, Son HS, Bae Y. Pkb/akt inhibits ceramide-induced apoptosis in neuroblastoma cells by blocking apoptosis-inducing factor (aif) translocation. J Cell Biochem. 102:1160-1170 2007;

22.

Lee JH, Won YS, Park KH, Lee MK, Tachibana H, Yamada K, Seo KI. Celastrol inhibits growth and induces apoptotic cell death in melanoma cells via the activation ros-dependent mitochondrial pathway and the suppression of pi3k/akt signaling. Apoptosis. 17:1275-1286 2012;

23.

Lima VN, Oliveira-Tintino CDM, Santos ES, Morais LP, Tintino SR, Freitas TS, Geraldo YS, Pereira RLS, Cruz RP, Menezes IRA, Coutinho HD. Antimicrobial and enhancement of the antibiotic activity by phenolic compounds: Gallic acid, caffeic acid and pyrogallol. Microb Pathog. 99:56-61 2016;

24.

Lin CH, Lu WC, Wang CW, Chan YC, Chen MK. Capsaicin induces cell cycle arrest and apoptosis in human kb cancer cells. BMC Complement Altern Med. 13:46 2013;

25.

Liu C, Boeren S, Miro Estruch I, Rietjens IMCM. The gut microbial metabolite pyrogallol is a more potent inducer of nrf2-associated gene expression than its parent compound green tea (-)-epigallocatechin gallate. Nutrients. 14:3392 2022;

26.

Lozano-Casabianca GA, Arango-Varela SS, Aguillon-Osma J, Llano-Ramirez MA, Maldonado-Celis ME. Inhibition of cell proliferation and induction of cell cycle arrest in colon cancer cells by lyophilized mango (Mangifera indica L.) pulp extract. Prev Nutr Food Sci. 27:436-447 2022;

27.

Ma YD, Thiyagarajan V, Tsai MJ, Lue SI, Chia YC, Shyue SK, Weng CF. Pyrogallol abates vsmc migration via modulation of caveolin-1, matrix metalloproteinase and intima hyperplasia in carotid ligation mouse. Environ Toxicol Pharmacol. 48:63-75 2016;

28.

Naeem A, Hu P, Yang M, Zhang J, Liu Y, Zhu W, Zheng Q. Natural products as anticancer agents: Current status and future perspectives. Molecules. 27:8367 2022;

29.

Nemec MJ, Kim H, Marciante AB, Barnes RC, Talcott ST, Mertens-Talcott SU. Pyrogallol, an absorbable microbial gallotannins-metabolite and mango polyphenols (Mangifera indica L.) suppress breast cancer ductal carcinoma in situ proliferation in vitro. Food Funct. 7:3825-3833 2016;

30.

Nicolis E, Lampronti I, Dechecchi MC, Borgatti M, Tamanini A, Bianchi N, Bezzerri V, Mancini I, Giri MG, Rizzotti P, Gambari R, Cabrini G. Pyrogallol, an active compound from the medicinal plant emblica officinalis, regulates expression of pro-inflammatory genes in bronchial epithelial cells. Int Immunopharmacol. 8:1672-1680 2008;

31.

Panaiyadiyan S, Kumar R. Prostate cancer nomograms and their application in asian men: A review. Prostate Int. 12:1-9 2024;

32.

Park WH, Park MN, Han YH, Kim SW. Pyrogallol inhibits the growth of gastric cancer snu-484 cells via induction of apoptosis. Int J Mol Med. 22:263-268 2008;

33.

Preya UH, Lee KT, Kim NJ, Lee JY, Jang DS, Choi JH. The natural terthiophene alpha-terthienylmethanol induces s phase cell cycle arrest of human ovarian cancer cells via the generation of ros stress. Chem Biol Interact. 272:72-79 2017;

34.

Ptak W, Rewicka M, Gryglewski A, Bielecka J. Differential effect of experimental diabetes on the early and late phase of contact sensitivity reaction in mice. Int Arch Allergy Appl Immunol. 81:136-140 1986;

35.

Pucci B, Kasten M, Giordano A. Cell cycle and apoptosis. Neoplasia. 2:291-299 2000;

36.

Rahman MA, Hannan MA, Dash R, Rahman MH, Islam R, Uddin MJ, Sohag AAM, Rahman MH, Rhim H. Phytochemicals as a complement to cancer chemotherapy: Pharmacological modulation of the autophagy-apoptosis pathway. Front Pharmacol. 12:639628 2021;

37.

Rodríguez-Berriguete G, Fraile B, Martínez-Onsurbe P, Olmedilla G, Paniagua R, Royuela M. Map kinases and prostate cancer. J Signal Transduc. 2012:169170 2012;

38.

Rodríguez-Berriguete G, Prieto A, Fraile B, Bouraoui Y, de Bethencourt FR, Martínez-Onsurbe P, Olmedilla G, Paniagua R, Royuela M. Relationship between il-6/erk and nf-κb: A study in normal and pathological human prostate gland. Eur Cytokine Netw. 21:241-250 2010;

39.

Roh T, Kim SW, Moon SH, Nam MJ. Genistein induces apoptosis by down-regulating thioredoxin-1 in human hepatocellular carcinoma snu-449 cells. Food Chem Toxicol. 97:127-134 2016;

40.

Saeki K, Hayakawa S, Isemura M, Miyase T. Importance of a pyrogallol-type structure in catechin compounds for apoptosis-inducing activity. Phytochemistry. 53:391-394 2000;

41.

Semprebon SC, Marques LA, D’epiro GFR, de Camargo EA, da Silva GN, Niwa AM, Macedo Junior F, Mantovani MS. Antiproliferative activity of goniothalamin enantiomers involves DNA damage, cell cycle arrest and apoptosis induction in mcf-7 and hb4a cells. Toxicol In Vitro. 30:250-263 2015;

42.

Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer J Clin. 74:12-49 2024;

43.

Sonpavde G, Kantoff PW. Immunotherapy for castration-resistant prostate cancer. Urol Clin North Am. 39:465-481 2012;

44.

Tanimoto T, Hori A, Kami M. Sipuleucel-t immunotherapy for castration-resistant prostate cancer. N Engl J Med. 363:411-422 2010;

45.

Vermeulen K, Van Bockstaele DR, Berneman ZN. The cell cycle: A review of regulation, deregulation and therapeutic targets in cancer. Cell Prolif. 36:131-149 2003;

46.

Walker AM, Stevens JJ, Ndebele K, Tchounwou PB. Evaluation of arsenic trioxide potential for lung cancer treatment: Assessment of apoptotic mechanisms and oxidative damage. J Cancer Sci Ther. 8:1-9 2016;

47.

Wang Q, Wei LW, Xiao HQ, Xue Y, Du SH, Liu YG, Xie XL. Methamphetamine induces hepatotoxicity via inhibiting cell division, arresting cell cycle and activating apoptosis: In vivo and in vitro studies. Food Chem Toxicol. 105:61-72 2017;

48.

Xu R, Hu J. The role of jnk in prostate cancer progression and therapeutic strategies. Biomed Pharmacother. 121:109679 2020;

49.

Yang CJ, Wang CS, Hung JY, Huang HW, Chia YC, Wang PH, Weng CF, Huang MS. Pyrogallol induces g2-m arrest in human lung cancer cells and inhibits tumor growth in an animal model. Lung Cancer. 66:162-168 2009;

50.

Yap WN, Chang P, Han HY, Lee DTW, Ling MT, Wong YC, Yap YL. Γ-tocotrienol suppresses prostate cancer cell proliferation and invasion through multiple-signalling pathways. Br J Cancer. 99:1832-1841 2008;

51.

Ye Y, Hou R, Chen J, Mo L, Zhang J, Huang Y, Mo Z. Formononetin-induced apoptosis of human prostate cancer cells through erk1/2 mitogen-activated protein kinase inactivation. Horm Metab Res. 44:263-267 2012;

Food Science and Preservation (FSP) reflected in Scopus

As of January 2024, the journal title has been changed to Food Science and Preservation (FSP).
We are pleased to announce that this has also been reflected in Scopus.
https://www.scopus.com/sourceid/21101210730
We look forward to your interest and submissions to the journal.
Thank you.

 


I don't want to open this window for a day.