Open Access

Ganoderma lucidum polysaccharide inhibits prostate cancer cell migration via the protein arginine methyltransferase 6 signaling pathway

  • Authors:
    • Xiaohui Zhao
    • Dayu Zhou
    • Yunen Liu
    • Chun Li
    • Xiaoguang Zhao
    • Ying Li
    • Wei Li
  • View Affiliations

  • Published online on: October 26, 2017     https://doi.org/10.3892/mmr.2017.7904
  • Pages: 147-157
  • Copyright: © Zhao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Prostate cancer is one of the most common types of malignant tumor of men worldwide and the incidence and mortality rate is gradually increasing. At present, the molecular mechanisms of growth and migration in human prostate cancer have not been completely elucidated. Studies have demonstrated that Ganoderma lucidum polysaccharides (GLP) can inhibit cancer. Therefore the present study investigated the effect and molecular mechanism of GLP on cell growth and migration of LNCaP human prostate cancer cells. LNCaP cells were transfected with either a protein arginine methyltransferase 6 (PRMT6) overexpression plasmid or PRMT6 small interfering (si)RNA. The cell growth and migration, and the expression of PRMT6 signaling‑associated proteins, were investigated following treatment with 5 and 20 µg/ml GLP. The results demonstrated that GLP inhibited cell growth, induced cell cycle arrest, decreased PRMT6, cyclin‑dependent kinase 2 (CDK2), focal adhesion kinase (FAK) and steroid receptor coactivator, (SRC) expression, and increased p21 expression in LNCaP cells, as determined by using a Coulter counter, flow cytometry, and reverse transcription‑quantitative polymerase chain reaction and western blotting, respectively. Furthermore, GLP significantly inhibited cell migration, as determined by Transwell migration and scratch assays, and altered CDK2, FAK, SRC and p21 expression in LNCaP cells transfected with the PRMT6 overexpression plasmid. By contrast, PRMT6 knockdown by siRNA reduced the effect of GLP on cell migration. These results indicate that GLP was effective in inhibiting cell growth, the cell cycle and cell migration, and the suppressive effect of GLP on cell migration may occur via the PRMT6 signaling pathway. Therefore, it is suggested that GLP may act as a tumor suppressor with applications in the treatment of prostate cancer. The results of the present study provide both the preliminary theoretical and experimental basis for the investigation of GLP as a therapeutic agent.

Introduction

Prostate cancer is one of the most common types of malignant tumor in Europe and the United States (1,2). Prostate cancer is the second most common non-skin cancer in men and is the fifth leading cause of cancer-associated mortality in men worldwide. A total of ~14% (1,22,000) of men diagnosed with prostate cancer worldwide in 2008 were in the Asia Pacific region, with 32% in Japan, 28% in China and 15% in Australia (3). It was recently reported that the incidence and mortality of prostate cancer in the majority of Asian countries gradually increased between 2012 and 2016 (4). It is established that prostate cancer is associated with genetic factors, diet, infection and hormonal factors. At present, the molecular mechanisms of growth and migration in human prostate cancer have not been completely elucidated.

Protein arginine methyltransferase 6 (PRMT6) is a type I arginine methyltransferase that is primarily expressed in the nucleus and has functions in the regulation of transcription and the cell cycle, and DNA repair (5). PRMT6 has also been demonstrated to act as a coactivator in estrogen, progesterone and glucocorticoid receptor transcription. Furthermore, El-Andaloussi et al (6) reported that PRMT6 had a key role in DNA base excision repair regulation as it forms a complex with methylated DNA polymerase β. Several studies have indicated that the expression of PRMT6 was usually observed in various types of tumor cells, including non-small cell lung cancer (7), hepatocellular carcinoma (8), breast cancer (9) and prostate cancer (10). In addition, studies have reported that PRMT6 knockdown inhibits cell growth and the cell cycle in lung cancer and U2OS human osteosarcoma cells (11,12). Phalke et al (13) reported that PRMT6 exhibited an oncogenic function by directly binding to and inhibiting the promoter of p21, which stimulated cell growth and protected the cell cycle from senescence in breast cancer cells. Although certain studies have reported that the expression of PRMT6 may be associated with the motility and invasion of tumor cells (10,14), the molecular mechanisms of PRMT6 in the regulation of cell growth and migration have not been completely elucidated.

It has been reported that Ganoderma lucidum (G. lucidum) exhibited preventive and therapeutic effects in cancer (15), chronic bronchitis (16), bronchial asthma (17) and hepatitis (18). Polysaccharides, which consist of glucose, mannose, galactose, xylose, fucose and arabinose, are one of the most important active components of G. lucidum (19). Several in vitro and in vivo studies have demonstrated that polysaccharides extracted from G. lucidum (GLP) exhibited significant effects on tumorigenesis, oxidative stress, inflammation and immunoregulation (20,21). Xu et al (22) reported that GLP affected the function of T lymphocytes, B lymphocytes, macrophages and natural killer cells. Although certain studies have reported that GLP exhibited potential antiproliferative, pro-apoptotic and inhibitory effects on migration in several cancer cell lines, including colon cancer (23,24), hepatocellular carcinoma (25), acute myeloid leukemia (26,27) and breast cancer (28,29), it is not established whether GLP is effective in regulating the growth and migration of prostate cancer cells. Therefore, the present study aimed to investigate the effect of GLP on the growth and migration of human prostate cancer cells, and to investigate the underlying molecular mechanism.

Materials and methods

Isolation and analysis of GLP

G. lucidum was provided by the College of Food Science of Shenyang Agricultural University (Shenyang, China). GLP was extracted from G. lucidum as described previously (30). To obtain a crude polysaccharide sample, fermentation broth of G. lucidum was concentrated and precipitated by 90% alcohol. Identification and quantification of the monosaccharides was performed using a gas chromatography (GC) analyzer (Beckman Coulter, Inc., Brea, CA, USA). According to the method described by He et al (31), 5 mg dry GLP was hydrolyzed in 2 M trifluoroacetic acid at 120°C for 5 h. The hydrolysate was reduced by NaBH4 and was acetylated using acetic anhydride. The acetylated monosaccharides of GLP were added into a GC analyzer and GLP was analyzed by gas chromatography, which determined that GLP was composed of arabinose, galactose, glucose and xylose in an approximate molar ratio of 4:2:10:1.

Cell culture and GLP treatment

LNCaP prostate cancer cells were purchased from ScienCell Research Laboratories, Inc., (San Diego, California, USA), and were cultured according to supplier instructions. Cells in culture bottle were washed three times with PBS and were cultured in RPMI-1640 (Thermo Fisher Scientific, Inc., Waltham, MA, USA), which was supplemented with 10% fetal calf serum (Thermo Fisher Scientific, Inc.), 2 mM glutamine, 100 U/ml penicillin and 100 mg/ml streptomycin. A total of 1.2×106−1.8×107 cells/well were subsequently treated with 5 and 20 µg/ml GLP and the control cells were treated with 0.01 M PBS (1XPBS) l at 37°C for 72 h. The concentrations of GLP employed were selected based on a previous study (32). All cells were cultured at 37°C, 5% CO2 and 100% humidity.

Plasmids and small interfering (si)RNA transfection

The PRMT6 expression plasmid pVAX1 (4 µg) acted as a control plasmid purchased from Thermo Fisher Scientific, Inc., PRMT6 siRNA (50 mM) and siR-Ribo (50 mM) acted as negative control were purchased from ScienCell Research Laboratories, Inc., which were transfected into LNCaP cells using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol at 37°C for 4 h. The following siRNA sequences were employed: PRMT6 siRNA, 5′-GAGCAAGACACGGACGUUU-3′. (GenBank, accession number BC073866). Cells following 4 h transfection were subsequently treated with 5 and 20 µg/ml GLP and the control cells were treated with 0.01 M PBS (1XPBS) at 37°C for 72 h.

Cell growth curve, cell clones and cycle analysis

For cell growth curve analysis, 5×104 cells were plated in each well of 12 well plates in triplicate. Cells were maintained in culture with 10% fetal calf serum at 37°C, 5% CO2 and 100% humidity for 12 h, and then cells were transfected with PRMT6 overexpression plasmid or PRMT6 siRNA for 4 h. Following transfection, 5×104 cells were treated with 5 or 20 µg/ml GLP and control cells were treated with 0.01 M PBS (1XPBS) at 37°C for 0, 24, 48, 72, 96 and 120 h and were immediately counted by a Coulter counter (Beckman Coulter, Inc.) through an optical microscope (Olympus Corporation, Tokyo, Japan) following staining with 0.4% trypan blue solution for 10 min at room temperature. A total of 5×104 cells/well were plated in 6-well plates in triplicate. Cells were maintained in culture with 10% fetal calf serum at 37°C, 5% CO2 and 100% humidity for 12 h, and then cells were transfected with the PRMT6 overexpression plasmid or PRMT6 siRNA for 4 h. Following transfection cells were treated with 5 or 20 µg/ml GLP and with 0.01 M PBS (1XPBS) as control at 37°C for 120 h, and then were fixed using 4% paraformaldehyde for 15 min at room temperature and stained with 0.4% trypan blue solution for 10 min at room temperature. Images were taken with an optical microscope (Olympus Corporation) magnification, ×100 and gray value analysis was using by imagepro version 6.0 (Media Cybernetics, Inc.) of five fields of view. For cell cycle analysis, 2×105 cells were fixed 70% pre-cooled ethanol, stored at 4°C overnight and then stained with PBS containing 5 mg/ml RNase, 0.1% Triton X-100 and 20 mg/ml propidium iodide in the dark at room temperature for 30 min and analyzed using a flow cytometer (Attune NxT; Thermo Fisher Scientific, Inc.). The amount of DNA in G1, S and G2/M phases was analyzed using ModFit 161 LT version 3.0 software (Verity Software House, Inc., Topsham, ME, USA).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was isolated using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and RT was performed using ABScript II cDNA First-Strand Synthesis kit (ABclonal Biotech Co., Ltd., Wuhan, China) according to the manufacturer's protocol. cDNA was quantified by qPCR using SYBR Premix Ex Taq II (Takara Bio, Inc., Otsu, Japan) on a Mx3000P qPCR system (Agilent, Santa Clara, CA, USA). The reaction was as follows: cDNA 2 µl, ddH2O 6.4 µl, upstream and downstream primers 0.8 µl, SYBR Premix Ex TaqTMII1 10 and 20 µl total reaction system. Amplification conditions: annealing 56°C, 30 sec; extending 72°C, 30 sec; 45 cycles. The experiment was repeated 3 times. Gene expression levels were calculated using Stratagene Mx3000P software (version Mx3005P; Shanghai PuDi Biotechnology Co., Ltd., Shanghai, China). The relative amount of PCR products generated from each primer set was determined on the basis of the threshold cycle (Ct) number using the 2−ΔΔCq method (33). GAPDH was used as control to normalize the amount of cDNA used. Relative expression=2[[Ct(control) gene X-Ct (treatment) gene X]-[Ct(control) 36B4-Ct(treatment) 36B4]]. The following primers were used: PRMT6, 5′-AGACACGGACGTTTCAGGAG-3′ (forward) and 5′-CCACTTTGTAGCGCAGCG-3′ (reverse); p21, 5′-TGAGCCGCGACTGTGATG-3′ (forward) and 5′-GTCTCGGTGACAAAGTCGAAGTT-3′ (reverse); cyclin-dependent kinase 2 (CDK2), 5′-ATGATGACGATGAGGGTGTGCCAA-3′ (forward) and 5′-GGTCACCATTGCAGCTGTCGAAAT-3′ (reverse); focal adhesion kinase (FAK), 5′-AGATGTACATCAAGGCATTTA-3′ (forward) and 5′-AATGCCTTGATGTACATCT-3′ (reverse); SRC, 5′-GCCTGTTCACCCGTACTCTGCC-3′ (forward) and 5′-GGTAACTGCCGATCATAAGGT-3′ (reverse); and GAPDH, 5′-AGAAGCTGGGGTCATTTG-3′ (forward) and 5′-AGGGGCCATCCACAGTCTTC-3′ (reverse).

Western blot analysis

Western blot analysis was performed as described previously (34). The cell protein was extracted by radioimmunoprecipitation lysate kit (Beijing Solarbio Science & Technology Co., Ltd., Beijing, China). The protein concentration was determined by bicinchoninic assay protein concentration kit (Vazyme, Piscataway, NJ, USA). Briefly, 20 µg protein/lane was loaded and resolved by 10% SDS-PAGE. Protein was subsequently transferred to polyvinylidene difluoride membranes. Following blocking with 5% bovine serum albumin (Thermo Fisher Scientific, Inc.) for 1 h at room temperature, primary antibodies against PRMT6 (1:500; cat. no. sc-271744; Santa Cruz Biotechnology, Inc., Dallas, TX, USA), p21 (1:500; cat. no. sc-136020; Santa Cruz Biotechnology, Inc.), CDK2 (1:1,000; cat. no. sc-70829; Santa Cruz Biotechnology, Inc.), FAK (1:200; cat. no. sc-271195; Santa Cruz Biotechnology, Inc.), SRC (1:1,000; cat. no. sc-32789; Santa Cruz Biotechnology, Inc.) and GAPDH (1:5,000; cat. no. sc-66163; Santa Cruz Biotechnology, Inc.) were incubated with the membranes at 4°C. After 12 h incubation, the blot was washed and incubated with goat anti-mouse immunoglobulin (Ig) G-horse radish peroxidase (HRP; 1:2,000; cat. no. sc-2005; Santa Cruz Biotechnology, Inc.) and goat anti-rabbit IgG-HRP (1:2,000; cat. no. sc-2004; Santa Cruz Biotechnology, Inc.) for 2.5 h at room temperature. Proteins were visualized using a Clarity Western enhanced chemiluminescence Substrate (Bio-Rad Laboratories, Inc., Hercules, CA, USA) and a Tanon 5200 Full automatic chemiluminescence image analysis system (Tanon Science and Technology Co., Ltd., Shanghai, China).

Transwell assay

Briefly, 5×105 cells were treated with 5 or 20 µg/ml GLP, or control treatment, for 24 h at 37°C and were seeded into the RPMI-1640 medium in the upper well of a Boyden chamber at a concentration of 5×104 cells per well. The lower compartment was filled with 1 ml RPMI-1640 (Thermo Fisher Scientific, Inc.), which was supplemented with 20% fetal calf serum (Thermo Fisher Scientific, Inc.). Following incubation at 37°C for 48 h, migratory cells on the lower surface of the filter were fixed with 95% ethanol for 10 min at room temperature, stained with 0.4% trypan blue solution for 10 min at room temperature and imaged using an optical microscope (Olympus Corporation) and an AxioCam HRc CCD camera (Olympus Corporation; magnification, ×400 times).

Scratch assay

Scratch assay was performed as described in a previous study (35). Briefly, 1×108 cells were plated into 6 well plates in triplicate at subconfluence and cultured for 24 h at 37°C. Confluent cells were treated either 5 or 20 µg/ml GLP, or PBS for the control treatment for 24 h at 37°C prior to cell scraping using 1 ml pipette tips. Cells were then washed with growth medium and continually cultured in RPMI-1640 (Thermo Fisher Scientific, Inc.), which was supplemented with 10% fetal calf serum (Thermo Fisher Scientific, Inc.), 2 mM glutamine, 100 U/ml penicillin and 100 mg/ml streptomycin for 48 h at 37°C, and during this time, cells were also treated either 5 or 20 µg/ml GLP, or PBS for the control treatment. Cell migration was photographed in ten regions at 0 and 48 h.

Statistical analysis

Data are presented as the mean ± standard deviation, and were analyzed by analysis of variance followed by Student-Newman-Keuls post-hoc test, and Chi-squared test using SPSS software (version 20.0; IBM Corp., Armonk, NY, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

GLP inhibits cell growth in LNCaP cells

The effects of different GLP concentrations on cell growth were determined using growth curve analyses and morphological observation. The results demonstrated that the growth ratio of cells was significantly decreased in either 5 or 20 µg/ml GLP groups compared with the control group following incubation for 72 h (Fig. 1A). Furthermore, morphological results also revealed that GLP (5 and 20 µg/ml) significantly inhibited cell growth compared with the control group (Fig. 1B-C).

GLP induces cell cycle arrest in LNCaP cells

Flow cytometry was performed to investigate the effect of GLP on the cell cycle. The results demonstrated that 5 and 20 µg/ml GLP significantly induced cell cycle arrest, with an increased number of cells in the G1 phase, of LNCaP cells in a dose-dependent manner, compared with control cells (Fig. 2).

GLP inhibits cell migration in LNCaP cells

Transwell and scratch assays were performed to investigate the effect of GLP on the migration of LNCaP cells. The results demonstrated that 5 and 20 µg/ml GLP significantly inhibited cell migration compared with the control group (Fig. 3).

Effect of GLP on the PRMT6 signaling pathway and migration-associated proteins in LNCaP cells

To determine the effect of GLP on the PRMT6 signaling pathway and migration-associated proteins in LNCaP cells, the expression of PRMT6, p21, CDK2, FAK and FRC were determined by RT-qPCR and western blot analysis. The results demonstrated that 5 and 20 µg/ml GLP decreased the protein expression of PRMT6 and CDK2, and increased p21 expression (Fig. 4A). In addition, the protein expression of migration-associated proteins FAK and FRC were decreased in GLP groups compared with the control group (Fig. 4A). Results for mRNA expression demonstrated that PRMT6, CDK2, FAK and FRC levels were significantly reduced at 5 and 20 µg/ml GLP, while p21 levels were significantly increased, compared with the control group (Fig. 4B-F).

GLP inhibits the migration of LNCaP cells via the PRMT6 signaling pathway

To determine whether GLP regulates the migration of LNCaP cells via the PRMT6 signaling pathway, cells were transfected with either a PRMT6 overexpression plasmid or PRMT6 siRNA, and were subsequently treated with 5 or 20 µg/ml GLP. The results demonstrated that overexpression of PRMT6 increased the expression of PRMT6, CDK2, FAK and FRC, and decreased p21 expression, compared with the vector-transfected control. By contrast, PRMT6 knockdown decreased PRMT6, CDK2, FAK and FRC expression, and increased p21 expression compared with the vector-transfected control. The results also demonstrated that GLP treatment inhibited PRMT6, CDK2, FAK and FRC expression, and promoted p21 expression in cells transfected with the PRMT6 overexpression plasmid, whereas PRMT6 knockdown by siRNA inhibited the effect of GLP on CDK2, p21, FAK and FRC expression (Fig. 5). In addition, Transwell results confirmed that GLP significantly inhibited the migration of cells transfected the PRMT6 overexpression plasmid, and results for PRMT6 siRNA-transfected cells indicated that PRMT6 siRNA prevented the effects of GLP on cell migration (Fig. 6).

Discussion

In recent years, the morbidity and mortality associated with prostate cancer has been increasing in China. The present study demonstrated that GLP inhibited the cell growth, cell cycle and cell migration, decreased PRMT6, CDK2, FAK and FRC expression, and increased p21 expression in LNCaP cells. Furthermore, the results indicated that GLP significantly inhibited cell migration and altered CDK2, FAK, FRC and p21 expression in cells transfected with a PRMT6 overexpression plasmid. By contrast, PRMT6 knockdown by siRNA reduced the effect of GLP on cell migration and CDK2, FAK, FRC and p21 expression.

Similar results were reported in a study by Ghafar et al (25), which demonstrated that GLP significantly inhibited the growth of hematoma cells and eliminated regulatory T cell suppression of T cell proliferation. Li et al (36) also reported that GLP decreased the adhesion of PC-3 M human prostate carcinoma cells to umbilical cord vascular endothelial cells. Furthermore, Liang et al (23) demonstrated that GLP exhibited potential antitumor activity by inhibiting migration and inducing apoptosis in human colon cancer cells. In addition, a previous study reported that Ganoderma atrum polysaccharide ameliorated the antitumor effect of cyclophosphamide, which was mediated via the induction of apoptosis and immune system activation in sarcoma 180-bearing mice (37). Collectively, the results of the current study indicated that GLP exhibited antitumor activity, which may partially be mediated by inhibiting the growth and migration of LNCaP cells.

PRMT6 expression is reported to be increased in various types of tumor cells, and it may participate in cell cycle regulation in tumor cells (3841). p21 is an important downstream gene of PRMT6 that is involved in the development of numerous tumor types, including osteosarcoma (42,43), liver cancer (44) and prostatic cancer (4547). In addition, p21 functions in the promotion G1 cell cycle arrest (48,49). CDK2 has important roles in several tumor types by modulating the migration and motility of cancer cells (50,51). FAK and FRC are key markers of tumor cell migration, which are closely associated with the development of certain tumor types, including hepatocellular carcinoma (52), breast cancer (53) and U87-MG glioma (54). In the current study, the results demonstrated that overexpression of PRMT6 significantly increased expression of PRMT6, CDK2, FAK and FRC, and decreased p21 expression. By contrast, PRMT6 knockdown significantly decreased PRMT6, CDK2, FAK and FRC expression, and increased p21 expression in LNCaP cells. A similar report indicated that PRMT6 knockdown significantly increased the expression of p21 and induced cell cycle arrest in breast cancer cells (13). In addition, Wang et al (55) demonstrated that PRMT6 overexpression reduced cell cycle arrest at G1 phase and decreased the intensity of p16-CDK4 association in A549 human lung adenocarcinoma cells. A previous study demonstrated that PRMT6 overexpression significantly decreased the cell growth and colony forming ability of MCF7 breast cancer cells compared with controls (14). It has also been reported that PRMT6 promoted the proliferation of U2OS human osteosarcoma cells and inhibited cell senescence by suppressing p21 expression (40,56). In conclusion, these results indicate that PRMT6 knockdown may inhibit cell migration by upregulating the expression of p21 and downregulating CDK2 expression in LNCaP cells.

However, it has been reported that GLP significantly inhibited the proliferation of S180 tumor-bearing mice by macrophage activation and improved immune system functions via the toll-like receptor 4-mediated nuclear factor-κB signaling pathway (57). The mitogen-activated protein kinase (MAPK) signaling pathway was also reported to be activated in GLP-induced RAW264.7 cells (58). In addition, a study by Liang et al (23) demonstrated that GLP inhibited migration and apoptosis by activating a Fas/caspase-dependent signaling pathway in human colon cancer cells. The results of the present study demonstrated that GLP significantly inhibited the migration of LNCaP cells transfected with a PRMT6 overexpression plasmid, whereas PRMT6 knockdown reduced the effect of GLP on cell migration, indicating that GLP may inhibit the migration of cells via the PRMT6 signaling pathway. A similar result was reported by Wu et al (59), which demonstrated that GLP inhibited the migration of MDA-MB-231 breast cancer cells, primarily by activating the FAK-Src signaling pathway. In addition, Yang et al (26) reported that GLP induced cell cycle arrest and apoptosis by blocking the extracellular signal-regulated kinase/MAPK pathway, and activating p38 and c-Jun N-terminal kinase MAPK pathways in HL-60 acute leukemia cells. WEES-G6, a triterpene-enriched extract from G. lucidum, was reported to inhibit the growth of Huh-7 human hepatoma cells, and activated JNK and p38 MAPK pathways in hepatocellular carcinoma (60).

In conclusion, the results of the present study indicated that GLP significantly inhibited the growth, cell cycle and migration of LNCaP cells. In addition, GLP inhibited the migration of cells transfected with a PRMT6 overexpression plasmid, whereas PRMT6 knockdown reduced the effect of GLP on cell migration, indicating that GLP may inhibit LNCaP cell migration via the PRMT6 signaling pathway. Therefore, it is suggested that GLP may act as a tumor suppressor have potential as a treatment for prostate cancer. The results of the present study provide both the preliminary theoretical and experimental basis for the investigation of GLP as a therapeutic agent.

Acknowledgements

The present study was supported by the Headmaster Foundation of Liaoning Medical University-Special Fund on Construction of Clinical Medicine (grant no. XZJJ20140233), the Science Technology Project in Liaoning Province (grant no. 20141173) and the National Natural Science Foundation of China (grant no. 11171042).

References

1 

Hashim D, Boffetta P, La Vecchia C, Rota M, Bertuccio P, Malvezzi M and Negri E: The global decrease in cancer mortality: Trends and disparities. Ann Oncol. 27:926–933. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Stegeman S, Amankwah E, Klein K, O'Mara TA, Kim D, Lin HY, Permuth-Wey J, Sellers TA, Srinivasan S, Eeles R, et al: A large-scale analysis of genetic variants within putative miRNA binding sites in prostate cancer. Cancer Discov. 5:368–379. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Baade PD, Youlden DR, Cramb SM, Dunn J and Gardiner RA: Epidemiology of prostate cancer in the Asia-Pacific region. Prostate Int. 1:47–58. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Monn MF, Tatem AJ and Cheng L: Prevalence and management of prostate cancer among East Asian men: Current trends and future perspectives. Urol Oncol. 34(58): e1–e9. 2016.

5 

Luo M, Li Y, Guo H, Lin S, Chen J, Ma Q, Gu Y, Jiang Z and Gui Y: Protein Arginine Methyltransferase 6 Involved in Germ Cell Viability during Spermatogenesis and Down-Regulated by the Androgen Receptor. Int J Mol Sci. 16:29467–29481. 2015. View Article : Google Scholar : PubMed/NCBI

6 

El-Andaloussi N, Valovka T, Toueille M, Steinacher R, Focke F, Gehrig P, Covic M, Hassa PO, Schär P, Hübscher U and Hottiger MO: Arginine methylation regulates DNA polymerase beta. Mol Cell. 22:51–62. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Ma WL, Wang L, Liu LX and Wang XL: Effect of phosphorylation and methylation on the function of the p16INK4a protein in non-small cell lung cancer A549 cells. Oncol Lett. 10:2277–2282. 2015.PubMed/NCBI

8 

Meerzaman DM, Yan C, Chen QR, Edmonson MN, Schaefer CF, Clifford RJ, Dunn BK, Dong L, Finney RP, Cultraro CM, et al: Genome-wide transcriptional sequencing identifies novel mutations in metabolic genes in human hepatocellular carcinoma. Cancer Genomics Proteomics. 11:1–12. 2014.PubMed/NCBI

9 

Dowhan DH, Harrison MJ, Eriksson NA, Bailey P, Pearen MA, Fuller PJ, Funder JW, Simpson ER, Leedman PJ, Tilley WD, et al: Protein arginine methyltransferase 6-dependent gene expression and splicing: Association with breast cancer outcomes. Endocr Relat Cancer. 19:509–526. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Vieira FQ, Costa-Pinheiro P, Ramalho-Carvalho J, Pereira A, Menezes FD, Antunes L, Carneiro I, Oliveira J, Henrique R and Jerónimo C: Deregulated expression of selected histone methylases and demethylases in prostate carcinoma. Endocr Relat Cancer. 21:51–61. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Kleinschmidt MA, de Graaf P, van Teeffelen HA and Timmers HT: Cell cycle regulation by the PRMT6 arginine methyltransferase through repression of cyclin-dependent kinase inhibitors. PLoS One. 7:e414462012. View Article : Google Scholar : PubMed/NCBI

12 

Yoshimatsu M, Toyokawa G, Hayami S, Unoki M, Tsunoda T, Field HI, Kelly JD, Neal DE, Maehara Y, Ponder BA, et al: Dysregulation of PRMT1 and PRMT6, Type I arginine methyltransferases, is involved in various types of human cancers. Int J Cancer. 128:562–573. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Phalke S, Mzoughi S, Bezzi M, Jennifer N, Mok WC, Low DH, Thike AA, Kuznetsov VA, Tan PH, Voorhoeve PM and Guccione E: p53-Independent regulation of p21Waf1/Cip1 expression and senescence by PRMT6. Nucleic Acids Res. 40:9534–9542. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Kim NH, Kim SN, Seo DW, Han JW and Kim YK: PRMT6 overexpression upregulates TSP-1 and downregulates MMPs: Its implication in motility and invasion. Biochem Biophys Res Commun. 432:60–65. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Gill BS, Navgeet and Kumar S: Ganoderma lucidum targeting lung cancer signaling: A review. Tumour Biol. 39:10104283177074372017. View Article : Google Scholar : PubMed/NCBI

16 

Batra P, Sharma AK and Khajuria R: Probing Lingzhi or Reishi medicinal mushroom Ganoderma lucidum (higher Basidiomycetes): A bitter mushroom with amazing health benefits. Int J Med Mushrooms. 15:127–143. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Liu C, Yang N, Song Y, Wang L, Zi J, Zhang S, Dunkin D, Busse P, Weir D, Tversky J, et al: Ganoderic acid C1 isolated from the anti-asthma formula, ASHMI™ suppresses TNF-α production by mouse macrophages and peripheral blood mononuclear cells from asthma patients. Int Immunopharmacol. 27:224–231. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Chiu HF, Fu HY, Lu YY, Han YC, Shen YC, Venkatakrishnan K, Golovinskaia O and Wang CK: Triterpenoids and polysaccharide peptides-enriched Ganoderma lucidum: A randomized, double-blind placebo-controlled crossover study of its antioxidation and hepatoprotective efficacy in healthy volunteers. Pharm Biol. 55:1041–1046. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Soccol CR, Bissoqui LY, Rodrigues C, Rubel R, Sella SR, Leifa F, de Souza Vandenberghe LP and Soccol VT: Pharmacological properties of biocompounds from spores of the lingzhi or reishi medicinal mushroom ganoderma lucidum (Agaricomycetes): A Review. Int J Med Mushrooms. 18:757–767. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Gokce EC, Kahveci R, Atanur OM, Gürer B, Aksoy N, Gokce A, Sargon MF, Cemil B, Erdogan B and Kahveci O: Neuroprotective effects of Ganoderma lucidum polysaccharides against traumatic spinal cord injury in rats. Injury. 46:2146–2155. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Liu YJ, Du JL, Cao LP, Jia R, Shen YJ, Zhao CY, Xu P and Yin GJ: Anti-inflammatory and hepatoprotective effects of Ganoderma lucidum polysaccharides on carbon tetrachloride-induced hepatocyte damage in common carp (Cyprinus carpio L.). Int Immunopharmacol. 25:112–120. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Xu Z, Chen X, Zhong Z, Chen L and Wang Y: Ganoderma lucidum polysaccharides: Immunomodulation and potential anti-tumor activities. Am J Chin Med. 39:15–27. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Liang ZE, Yi YJ, Guo YT, Wang RC, Hu QL and Xiong XY: Inhibition of migration and induction of apoptosis in LoVo human colon cancer cells by polysaccharides from Ganoderma lucidum. Mol Med Rep. 12:7629–7636. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Liang Z, Guo YT, Yi YJ, Wang RC, Hu QL and Xiong XY: Ganoderma lucidum polysaccharides target a Fas/caspase dependent pathway to induce apoptosis in human colon cancer cells. Asian Pac J Cancer Prev. 15:3981–3996. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Li A, Shuai X, Jia Z, Li H, Liang X, Su D and Guo W: Ganoderma lucidum polysaccharide extract inhibits hepatocellular carcinoma growth by downregulating regulatory T cells accumulation and function by inducing microRNA-125b. J Transl Med. 13:1002015. View Article : Google Scholar : PubMed/NCBI

26 

Yang G, Yang L, Zhuang Y, Qian X and Shen Y: Ganoderma lucidum polysaccharide exerts anti-tumor activity via MAPK pathways in HL-60 acute leukemia cells. J Recept Signal Transduct Res. 36:6–13. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Chui CH, Wong RS, Cheng GY, Lau FY, Kok SH, Cheng CH, Cheung F, Tang WK, Teo IT, Chan AS and Tang JC: Antiproliferative ability of a combination regimen of crocodile egg extract, wild radix ginseng and natural Ganoderma lucidum on acute myelogenous leukemia. Oncol Rep. 16:1313–1316. 2006.PubMed/NCBI

28 

Zhang Y: Ganoderma lucidum (Reishi) suppresses proliferation and migration of breast cancer cells via inhibiting Wnt/β-catenin signaling. Biochem Biophys Res Commun. 488:679–684. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Smina TP, Nitha B, Devasagayam TP and Janardhanan KK: Ganoderma lucidum total triterpenes induce apoptosis in MCF-7 cells and attenuate DMBA induced mammary and skin carcinomas in experimental animals. Mutat Res. 813:45–51. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Huang JH, Ailati A and Mao J: Purification and Structural Identification of a Bioactive Polysaccharide Fraction from Ganoderma lucidum. Food Sci. 32:301–304. 2011.

31 

He Y, Ye M, Du Z, Wang H, Wu Y and Yang L: Purification, characterization and promoting effect on wound healing of an exopolysaccharide from Lachnum YM405. Carbohydr Polym. 105:169–176. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Shang D, Li Y, Wang C, Wang X, Yu Z and Fu X: A novel polysaccharide from Se-enriched Ganoderma lucidum induces apoptosis of human breast cancer cells. Oncol Rep. 25:267–272. 2011.PubMed/NCBI

33 

Livak KJ and Schmittgen TD: Analysis of Relative Gene Expression Data Using RealTime Quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

34 

Kim SY, Seo M, Kim Y, Lee YI, Oh JM, Cho EA, Kang JS and Juhnn YS: Stimulatory heterotrimeric GTP-binding protein inhibits hydrogen peroxide-induced apoptosis by repressing BAK induction in SH-SY5Y human neuroblastoma cells. J Biol Chem. 283:1350–1361. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Jing H, Liaw L, Friesel R, Vary C, Hua S and Yang X: Suppression of Spry4 enhances cancer stem cell properties of human MDA-MB-231 breast carcinoma cells. Cancer Cell Int. 16:192016. View Article : Google Scholar : PubMed/NCBI

36 

Li YB, Wang R, Wu HL, Li YH, Zhong LJ, Yu HM and Li XJ: Serum amyloid A mediates the inhibitory effect of Ganoderma lucidum polysaccharides on tumor cell adhesion to endothelial cells. Oncol Rep. 20:549–556. 2008.PubMed/NCBI

37 

Li W, Nie S, Chen Y, Wang Y, Li C and Xie M: Enhancement of cyclophosphamide-induced antitumor effect by a novel polysaccharide from Ganoderma atrum in sarcoma 180-bearing mice. J Agric Food Chem. 59:3707–3716. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Kim C, Lim Y, Yoo BC, Won NH, Kim S and Kim G: Regulation of post-translational protein arginine methylation during HeLa cell cycle. Biochim Biophys Acta. 1800:977–985. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Stein C, Nötzold RR, Riedl S, Bouchard C and Bauer UM: The arginine methyltransferase PRMT6 cooperates with polycomb proteins in regulating HOXA Gene Expression. PLoS One. 11:e01488922016. View Article : Google Scholar : PubMed/NCBI

40 

Stein C, Riedl S, Ruthnick D, Nötzold RR and Bauer UM: The arginine methyltransferase PRMT6 regulates cell proliferation and senescence through transcriptional repression of tumor suppressor genes. Nucleic Acids Res. 40:9522–9533. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Neault M, Mallette FA, Vogel G, Michaud-Levesque J and Richard S: Ablation of PRMT6 reveals a role as a negative transcriptional regulator of the p53 tumor suppressor. Nucleic Acids Res. 40:9513–9521. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Lv H, Gao G, Zhang L and Sun Y: Pololike kinase 3 inhibits osteosarcoma cell proliferation and tumorigenesis via cooperative interaction with p21. Mol Med Rep. 12:6789–6796. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Wan D, Jiang C, Hua X, Wang T and Chai Y: Cell cycle arrest and apoptosis induced by aspidin PB through the p53/p21 and mitochondria-dependent pathways in human osteosarcoma cells. Anticancer Drugs. 26:931–941. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Wang J, Lei ZJ, Guo Y, Wang T, Qin ZY, Xiao HL, Fan LL, Chen DF, Bian XW, Liu J and Wang B: miRNA-regulated delivery of lincRNA-p21 suppresses β-catenin signaling and tumorigenicity of colorectal cancer stem cells. Oncotarget. 6:37852–37870. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Al-Azayzih A, Gao F and Somanath PR: P21 activated kinase-1 mediates transforming growth factor β1-induced prostate cancer cell epithelial to mesenchymal transition. Biochim Biophys Acta. 1853:1229–1239. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Di Giacomo V, Di Valerio V, Rapino M, Bosco D, Cacciatore I, Ciulla M, Marrazzo A, Fiorito J, Di Stefano A and Cataldi A: MRJF4, a novel histone deacetylase inhibitor, induces p21 mediated autophagy in PC3 prostate cancer cells. Cell Mol Biol (Noisy-le-grand). 61:17–23. 2015.PubMed/NCBI

47 

Isin M, Uysaler E, özgur E, Koseoglu H, Sanli ö, Yucel öB, Gezer U and Dalay N: Exosomal lncRNA-p21 levels may help to distinguish prostate cancer from benign disease. Front Genet. 6:1682015.PubMed/NCBI

48 

Kim SH, Hwang KA, Shim SM and Choi KC: Growth and migration of LNCaP prostate cancer cells are promoted by triclosan and benzophenone-1 via an androgen receptor signaling pathway. Environ Toxicol Pharmacol. 39:568–576. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Lin HP, Lin CY, Huo C, Hsiao PH, Su LC, Jiang SS, Chan TM, Chang CH, Chen LT, Kung HJ, et al: Caffeic acid phenethyl ester induced cell cycle arrest and growth inhibition in androgen-independent prostate cancer cells via regulation of Skp2, p53, p21Cip1 and p27Kip1. Oncotarget. 6:6684–6707. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Wei K, Ye Z, Li Z, Dang Y, Chen X, Huang N, Bao C, Gan T, Yang L and Chen G: An immunohistochemical study of cyclin-dependent kinase 5 (CDK5) expression in non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC): A possible prognostic biomarker. World J Surg Oncol. 14:342016. View Article : Google Scholar : PubMed/NCBI

51 

Bisht S, Nolting J, Schutte U, Haarmann J, Jain P, Shah D, Brossart P, Flaherty P and Feldmann G: Cyclin-Dependent Kinase 5 (CDK5) controls melanoma cell motility, invasiveness and metastatic spread-identification of a promising novel therapeutic target. Transl Oncol. 8:295–307. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Chen JS, Li HS, Huang JQ, Dong SH, Huang ZJ, Yi W, Zhan GF, Feng JT, Sun JC and Huang XH: MicroRNA-379-5p inhibits tumor invasion and metastasis by targeting FAK/AKT signaling in hepatocellular carcinoma. Cancer Lett. 375:73–83. 2016. View Article : Google Scholar : PubMed/NCBI

53 

Sanchez Castillo R, Gomez R and Salazar Perez E: Bisphenol A Induces Migration through a GPER-, FAK-, Src- and ERK2-Dependent Pathway in MDA-MB-231 Breast Cancer Cells. Chem Res Toxicol. 29:285–295. 2016. View Article : Google Scholar : PubMed/NCBI

54 

Li S, Li C, Ryu HH, Lim SH, Jang WY and Jung S: Bacitracin inhibits the migration of U87-MG glioma cells via interferences of the integrin outside-in signaling pathway. J Korean Neurosurg Soc. 59:106–116. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Wang X, Huang Y, Zhao J, Zhang Y, Lu J and Huang B: Suppression of PRMT6-mediated arginine methylation of p16 protein potentiates its ability to arrest A549 cell proliferation. Int J Biochem Cell Biol. 44:2333–2341. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Michaud-Levesque J and Richard S: Thrombospondin-1 is a transcriptional repression target of PRMT6. J Biol Chem. 284:21338–21346. 2009. View Article : Google Scholar : PubMed/NCBI

57 

Zhang S, Nie S, Huang D, Huang J, Wang Y and Xie M: Polysaccharide from Ganoderma atrum evokes antitumor activity via Toll-like receptor 4-mediated NF-κB and mitogen-activated protein kinase signaling pathways. J Agric Food Chem. 61:3676–3682. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Yu Q, Nie SP, Wang JQ, Yin PF, Li WJ and Xie MY: Polysaccharide from Ganoderma atrum induces tumor necrosis factor-α secretion via phosphoinositide 3-kinase/Akt, mitogen-activated protein kinase and nuclear factor-κB signaling pathways in RAW264.7 cells. Int Immunopharmacol. 14:362–368. 2012. View Article : Google Scholar : PubMed/NCBI

59 

Wu GS, Song YL, Yin ZQ, Guo JJ, Wang SP, Zhao WW, Chen XP, Zhang QW, Lu JJ and Wang YT: Ganoderiol A-enriched extract suppresses migration and adhesion of MDA-MB-231 cells by inhibiting FAK-SRC-paxillin cascade pathway. PLoS One. 8:e766202013. View Article : Google Scholar : PubMed/NCBI

60 

Lin SB, Li CH, Lee SS and Kan LS: Triterpene-enriched extracts from Ganoderma lucidum inhibit growth of hepatoma cells via suppressing protein kinase C, activating mitogen-activated protein kinases and G2-phase cell cycle arrest. Life Sci. 72:2381–2390. 2003. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2018
Volume 17 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao X, Zhou D, Liu Y, Li C, Zhao X, Li Y and Li W: Ganoderma lucidum polysaccharide inhibits prostate cancer cell migration via the protein arginine methyltransferase 6 signaling pathway. Mol Med Rep 17: 147-157, 2018
APA
Zhao, X., Zhou, D., Liu, Y., Li, C., Zhao, X., Li, Y., & Li, W. (2018). Ganoderma lucidum polysaccharide inhibits prostate cancer cell migration via the protein arginine methyltransferase 6 signaling pathway. Molecular Medicine Reports, 17, 147-157. https://doi.org/10.3892/mmr.2017.7904
MLA
Zhao, X., Zhou, D., Liu, Y., Li, C., Zhao, X., Li, Y., Li, W."Ganoderma lucidum polysaccharide inhibits prostate cancer cell migration via the protein arginine methyltransferase 6 signaling pathway". Molecular Medicine Reports 17.1 (2018): 147-157.
Chicago
Zhao, X., Zhou, D., Liu, Y., Li, C., Zhao, X., Li, Y., Li, W."Ganoderma lucidum polysaccharide inhibits prostate cancer cell migration via the protein arginine methyltransferase 6 signaling pathway". Molecular Medicine Reports 17, no. 1 (2018): 147-157. https://doi.org/10.3892/mmr.2017.7904