Huaier suppresses proliferative and metastatic potential of prostate cancer PC3 cells via downregulation of Lamin B1 and induction of autophagy

  • Authors:
    • Ailin Yang
    • Yanan Zhao
    • Ying Wang
    • Xiaojun Zha
    • Yunfang Zhao
    • Pengfei Tu
    • Zhongdong Hu
  • View Affiliations

  • Published online on: April 5, 2018     https://doi.org/10.3892/or.2018.6358
  • Pages: 3055-3063
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 malignancies occurring in males. Although large advances have been made in the pathogenesis of prostate cancer, the development of drugs with high efficacy and low toxicity for the treatment of prostate cancer is urgently needed. Recently, more and more attention has been paid to the antitumor effect of Traditional Chinese Medicine (TCM) worldwide. Trametes robiniophila Murr. (Huaier) has been applied as a type of TCM drug for ~1,600 years. Huaier exhibits excellent clinical efficacy in the treatment of cancer, including prostate cancer. However, the mechanisms underlying the anti-prostate cancer effect of Huaier remain largely unclear. In the present study, we revealed that Huaier aqueous extract inhibited the proliferative and metastatic capabilities of human prostate cancer PC3 cells through CCK-8 assay, in vitro scratch assay and Transwell assay. Moreover, decreased Lamin B1 was implicated in Huaier-induced suppression of proliferative and metastatic potential of PC3 cells. Intriguingly, we demonstrated that Huaier treatment induced autophagic cell death in PC3 cells. This study sheds new light on the mechanisms underlying the activity of Huaier against prostate cancer and provides a new theoretical basis for the clinical application of Huaier in prostate cancer.

Introduction

Prostate cancer is one of the leading causes of cancer-related death in men worldwide. At present, treatment approaches for prostate cancer mainly include surgery, chemotherapy and hormonal therapy (1,2). The selection of the treatment method for prostate cancer generally depends on the clinical stage, Gleason score, age of patients, and other factors. To date, the chemotherapeutic effect for prostate cancer is limited due to drug resistance and cytotoxicity (35), which makes it urgent to develop new therapeutic drugs to further improve the clinical outcomes of patients with prostate cancer.

Recently, increased attention has been paid to the anticancer effects of Traditional Chinese Medicine (TCM) largely due to the advantages of low toxicity and multi-targets (68). Trametes robiniophila Murr. (Huaier) has a long history of disease treatment for more than 1,600 years in China. Numerous studies have revealed that Huaier exhibits superior effects for the treatment of several types of cancers, such as hepatocellular carcinoma, breast, ovarian, lung and prostate cancer (914). The underlying mechanisms of the anticancer effects of Huaier include the inhibition of tumor cell proliferation, metastasis and angiogenesis, as well as induction of apoptosis (11,13,1517). However, the mechanisms underlying the anti-prostate cancer effect of Huaier remain to be elucidated.

In the present study, we demonstrated that Huaier significantly inhibited the proliferative and metastatic potential of human prostate cancer PC3 cells. Moreover, downregulation of Lamin B1 was responsible for the inhibition of the proliferative and metastatic capacity of PC3 cells exposed to Huaier aqueous extract. More importantly, Huaier treatment activated autophagy in PC3 cells, and suppression of autophagy attenuated Huaier-induced cell death in PC3 cells. Thus, the present study provides a theoretical and experimental basis for the clinical application of Huaier for the treatment of prostate cancer.

Materials and methods

Reagents and antibodies

Ham's F-12K (Kaighn's) medium, fetal bovine serum (FBS), penicillin-streptomycin solution, 0.25% trypsin, Matrigel and Transwell chambers were purchased from Corning Life Sciences (Corning, NY, USA). Cell Counting Kit-8 (CCK-8) was obtained from Dojindo Laboratories (Kumamoto, Japan). 3-Methyladenine (3-MA) was purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Bafilomycin A1 was obtained from Aladdin Shanghai Biochemical Technology Co., Ltd. (Shanghai, China). Bis-Tris Nu-PAGE gels (4–12%) were obtained from Invitrogen (Thermo Fisher Scientific, Inc., Waltham, MA, USA). Super ECL Plus was from GE Healthcare (Pittsburgh, PA, USA). β-actin antibody was purchased from Abgent (cat. no. AM1829b; San Diego, CA, USA). Atg3 (cat. no. 3415), Beclin-1 (cat. no. 3495) and Atg5 (cat. no. 12994) antibodies were purchased from Cell Signaling Technology (Danvers, MA, USA). LC3 antibody was from MBL (cat. no. PM036; Tokyo, Japan). Lamin B1 (cat. no. sc-20682), HRP-labeled goat anti-mouse (cat. no. sc-2005) and goat anti-rabbit (cat. no. sc-2004) antibodies were from Santa Cruz Biotechnology.

Preparation of Huaier aqueous extract

The electuary ointment of Huaier was obtained from Gaitianli Medicine Co., Ltd. (Jiangsu, China). It was dissolved in F-12K complete medium to obtain a 10 mg/ml stock solution and was stored at 4°C after sterilization by filtration.

Cell culture

Human prostate cancer PC3 cells were obtained from the Cell Culture Center of the Institute of Basic Medical Sciences of the Chinese Academy of Medical Sciences (Beijing, China). PC3 cells were maintained in Ham's F-12K (Kaighn's) medium containing 10% FBS and 1% penicillin/streptomycin at 37°C and 5% CO2.

Cell viability assay

PC3 cells were seeded into 96-well plates at a density of 3,500 cells/well. On the following day, the cells were treated with Huaier aqueous extract at the indicated concentrations for different times. Afterwards, 10 µl of CCK-8 was added into each well and incubated at 37°C for 2 h. The optical density (OD) was then measured at 450 nm using a microplate reader (Perkin-Elmer, Waltham, MA, USA).

In vitro scratch assay

The scratch assay was performed as described previously (10,18). In brief, PC3 cells were seeded into 12-well plates with complete medium. Cells in a subconfluent state were starved with serum-free medium for 12 h, and then a straight cell-free wound was created using a 10-µl pipette tip. Next, the cells were maintained in serum-free medium containing 4 mg/ml of Huaier extract. The scratch width was measured at 0 and 24 h. The cell migration distances were analyzed quantitatively.

Transwell assay

The Transwell system was established as previously described (19,20). PC3 cells (1×105) were resuspended in 200 µl serum-free medium containing Huaier extract (4 mg/ml) and then added to the upper chamber of the Transwell system. The lower chamber was filled with 750 µl complete medium containing 10% FBS. After incubation for 36 h, the cells were removed from the upper surface of the membrane with a cotton swab. Next, the cells on the bottom surface of the membrane were fixed and then stained with crystal violet. The Transwell chamber was washed twice with phosphate-buffered saline (PBS) to remove the dye. The invasive cells were observed and counted on 5 random fields under an inverted microscope (Leica Microsystems GmbH, Wetzlar, Germany).

Quantitative real-time PCR

Total RNA was extracted from PC3 cells using E.Z.N.A.® Total RNA Kit I (Omega Bio-tek, Inc., Norcross, GA, USA) and reverse-transcribed into cDNA using the PrimeScript RT reagent kit (Takara Biotechnology, Inc., Dalian, China) according to the manufacturer's protocols. cDNA was used as a template for the quantitative PCR using the TransStart Top Green qPCR SuperMix (Beijing TransGen Biotech Co., Ltd., Beijing, China). The primer sequences were: human Lamin B1 forward, 5′-TTCTCGAAGCTTGATCTGGG-3′ and human Lamin B1 reverse, 5′-GATCGAGCTGGGCAAGTG-3′; human β-actin forward, 5′-GTTGTCGACGACGAGCG-3′ and human β-actin reverse, 5′-GCACAGAGCCTCGCCTT-3′.

Immunoblotting

Cells were washed twice with PBS and harvested with lysis buffer [10 mM Tris (pH 6.8), 2% SDS, 10% glycerol and 100 mM DTT], and then cell lysates were boiled for 10 min at 98°C. The levels of indicated proteins were detected by immunoblot analysis as previously described (21).

RNA interference

All siRNAs were synthesized by Shanghai GenePharma Co., Ltd. (Shanghai, China). PC3 cells were seeded in 6-well plates and then transfected with siRNAs targeting Lamin B1, Atg5 or Beclin-1 using Lipofectamine 2000 (Thermo Fisher Scientific, Inc., Waltham, MA, USA) according to the manufacturer's instructions. The siRNA target sequences were: Lamin B1 (human), 5′-CGCGCTTGGTAGAGGTGGA-3′; Atg5 (human), 5′-GGACGAATTCCAACTTGTT-3′; Beclin-1 (human), 5′-CAGTTTGGCACAATCAATA-3′; negative control (NC), 5′-TTCTCCGAACGTGTCACGT-3′.

Electron microscopy

Cells were collected with 2.5% glutaraldehyde, and then centrifuged (1,000 revolutions, 10 min) and washed twice with PBS. The samples were post-fixed with 1% osmium tetroxide at 4°C for 2 h in the dark and then washed three times with 0.1MPB. After dehydration, the permeation, paraffin embedding and section staining were performed as previously described (22). The ultrastructure of cells was observed under a JEM-1230 transmission electron microscope (JEOL, Ltd., Tokyo, Japan).

Acridine orange staining

PC3 cells treated with 8 mg/ml Huaier exact for indicated times were washed twice with PBS and stained with 10 µg/ml acridine orange for 20 min at 37°C in the dark. The cells were observed under an inverted fluorescence microscope.

Immunofluorescence assay

Cells seeded on confocal dishes were washed with PBS and fixed in 4% paraformaldehyde for 10 min, and then permeabilized with PBS containing 0.5% Triton X-100 for 20 min. Next, the cells were washed three times with PBS and blocked with 1% BSA for 1 h at 37°C, and then incubated with the primary antibody (1:100) overnight at 4°C. Subsequently, the cells were washed with PBS and then incubated with anti-rabbit FITC-conjugated secondary antibody (1:100) for 1 h at 37°C. Nucleus was counterstained with DAPI in the dark after being washed with PBS. The cells were observed and photographed with a laser scanning confocal microscope (Olympus FV1000; Olympus Corp., Tokyo, Japan).

Statistical analysis

The data are presented as mean ± SD of triplicate samples. The statistical analysis in this study was evaluated by the Student's t-test and ANOVA using GraphPad Prism 5.0 software (GraphPad Software, Inc., La Jolla, CA, USA). P<0.05 was considered to indicate a statistically significant result.

Results

Huaier inhibits the proliferative and metastatic potential of human prostate cancer PC3 cells

CCK-8 assay indicated that Huaier aqueous extract significantly inhibited proliferation of the PC3 cells in a time- and dose-dependent manner (Fig. 1A). The IC50 value of PC3 cells exposed to Huaier for 48 h was 8.18 mg/ml. The wound healing assay is widely used to evaluate the migratory ability of cells in vitro. As shown in Fig. 1B, the migration capacity of PC3 cells was decreased after treatment with Huaier, and the migration inhibition rate of PC3 cells treated with Huaier was 44.68±16.39%. Moreover, the invasion capacity of cells in vitro is frequently examined by using Transwell assay. As shown in Fig. 1C, after treatment of PC3 cells with 4 mg/ml Huaier aqueous extract for 36 h, the number of cells that had successfully passed through the Matrigel-coated membrane was markedly reduced compared with that of the control group cells. Taken together, Huaier markedly suppressed the proliferative and metastatic capability of the human prostate cancer cells.

Huaier downregulates Lamin B1 expression in human prostate cancer PC3 cells

Lamin B1 is a member of the lamin family making up the nuclear matrix and is abnormally overexpressed in multiple types of cancers including prostate, hepatocellular carcinoma and pancreatic cancer (19,2325). Moreover, Lamin B1 positively regulates the proliferation and invasion of pancreatic cancer cells (25). Our previous study revealed that as a pro-oncogenic gene, Lamin B1 was dramatically decreased in human hepatoma SKHEP-1 cells exposed to Huaier (10). Intriguingly, qRT-PCR and western blot analysis demonstrated that Huaier aqueous extract significantly inhibited the mRNA and protein levels of Lamin B1 in PC3 cells in a dose-dependent manner (Fig. 2A and B).

Lamin B1 is involved in the inhibition of proliferation and metastatic potential of PC3 cells exposed to Huaier

Next, we investigated whether downregulation of Lamin B1 contributes to Huaier-mediated inhibition of proliferative and metastatic capacity of PC3 cells by using RNA interference. Transfection with siRNAs targeting Lamin B1 markedly reduced Lamin B1 expression (Fig. 3A). Moreover, depletion of Lamin B1 significantly blunted the proliferation of PC3 cells (Fig. 3B). In addition, reduction of Lamin B1 substantially suppressed the migration and invasion of PC3 cells (Fig. 3C and D). Thus, Lamin B1 positively regulates the proliferation and metastatic potential of human prostate cancer PC3 cells. To further investigate the role of Lamin B1 in the inhibition of proliferative and metastatic potential of PC3 cells in the presence of Huaier, we treated PC3 cells transfected with Lamin B1 siRNAs or negative control siRNAs with Huaier aqueous extract. As depicted in Fig. 4A, depletion of Lamin B1 attenuated the inhibitory effect of Huaier on the proliferation of PC3 cells. Additionally, the suppression of migration and invasion of PC3 cells caused by Huaier treatment were significantly impaired by knockdown of Lamin B1 (Fig. 4B and C). Collectively, decreased Lamin B1 is partially responsible for the inhibition of proliferation and the metastatic potential of PC3 cells in the presence of Huaier.

Huaier induces autophagy in PC3 cells

Autophagy is one of the underlying mechanisms for drug-induced cell death (26,27). In the present study we determined the effect of Huaier on autophagy in PC3 cells. Observation of autophagosomes and other related subcellular structures in the cytoplasm with transmission electron microscope is widely used for autophagy assessment (22,28,29). Ultrastructural changes of PC3 cells treated with Huaier were observed by transmission electron microscope. As shown in Fig. 5A, PC3 cells treated with Huaier exhibited typical characteristics of autophagy: autophagosomes and autolysosomes which contained cytoplasmic components. Moreover, acridine orange staining was used to observe the accumulation of autophagy vesicles. The number of acidic vesicles marked by orange fluorescence increased in response to Huaier treatment in a time-dependent manner (Fig. 5B). A series of autophagy-related proteins participate in different stages of autophagosome formation, such as Atg3, Atg5 and Beclin-1 (3033). LC3 is an autophagosomal marker protein (34). LC3-II, one form of LC3, accumulates on the membranes of autophagosomes and is widely used as a marker for autophagy evaluation (34,35). Western blot analysis revealed that Huaier treatment led to increase in the expression of Atg3, Atg5, Beclin-1 and LC3-II in a dose- and time-dependent manner in PC3 cells (Fig. 5C and D). Moreover, Huaier treatment markedly increased LC3 puncta in number and intensity in PC3 cells under a fluorescence microscope (Fig. 5E), indicating that the number of autophagosomes was increased in the presence of Huaier. Taken together, Huaier evidently promoted autophagy in PC3 cells.

Autophagy inhibition attenuates Huaier-induced inhibition of proliferation of PC3 cells

To investigate whether autophagy was involved in the inhibition of proliferation of PC3 cells by Huaier, we suppressed autophagy with two autophagy inhibitors and RNA interference. As depicted in Fig. 6A and B, inhibition of autophagy with 3-MA or bafilomycin A1 significantly impaired the sensitivity of PC3 cells to Huaier treatment. Moreover, Atg5 and Beclin-1, two essential components involved in autophagosome formation (36), were efficiently knocked down in the PC3 cells transfected with siRNAs (Fig. 6C and D). Consistent with the results of autophagy inhibitors, suppression of autophagy with siRNAs against Atg5 or Beclin-1 remarkably blunted the inhibitory effect of Huaier on the proliferation of PC3 cells (Fig. 6C and D). Collectively, the inhibitory proliferation of PC3 cells exposed to Huaier is partially mediated by activation of autophagy.

Discussion

In recent years, the antitumor effects of Huaier have drawn the attention of cancer researchers. Many clinical applications have demonstrated that Huaier can be used for the treatment of multiple types of cancers including prostate cancer. However, the anti-prostate cancer effect of Huaier and its underlying mechanisms remain elusive. In the present study, we demonstrated that Huaier inhibited the proliferation and metastatic potential of human prostate cancer PC3 cells partially through downregulation of Lamin B1. In addition, we revealed that Huaier treatment induced autophagic cell death in PC3 cells.

Uncontrolled cell proliferation is one of the hallmarks of cancer (37). Inhibition of cancer cell proliferation can be used as a strategy to treat cancer. Metastasis is a complex multi-step process which plays a pivotal role in the progression of cancer (38). Cancer metastasis is responsible for ~90% of human cancer-related deaths (39). Metastases to distant organs such as the bone, liver, lungs and brain frequently occur in the advanced stage of prostate cancer (40,41). Most prostate cancer-related deaths result from metastases. Herein we revealed that Huaier had excellent anti-proliferative and anti-metastatic effects in PC3 cells. Thus, Huaier can be used as a candidate drug for targeting the proliferation and metastasis of cancer cells in the treatment of human prostate cancer. Lamin B1 is an important member of the lamin protein family (23) and was reported to be a carcinogenic gene (25). The levels of Lamin B1 are elevated in tumors of patients with hepatocellular carcinoma (24). Moreover, Lamin B1 expression was found to be markedly increased in malignant prostate cancer (19). Knockdown of Lamin B1 expression by siRNAs induced apoptosis in HeLa cells (42). Our previous study demonstrated that Lamin B1 was remarkably downregulated in human hepatoma SKHEP-1 cells exposed to Huaier (10). In the present study, we revealed that Huaier treatment markedly reduced Lamin B1 expression in human prostate cancer PC3 cells. Furthermore, Lamin B1 was required for the inhibition of proliferation and metastatic potential of PC3 cells by Huaier. As an oncogenic protein, Lamin B1 is a novel target of Huaier for prostate cancer treatment.

Autophagy is a normal physiological process in which the cytoplasmic components including misfolded proteins and damaged organelles are surrounded to form autophagosomes that are eventually transported to lysosomes for degradation (20,43). As one of the underlying mechanisms of cell death, autophagy frequently occurs in cancer cells in response to antitumor therapies (4447). Huaier triggered autophagy in human breast cancer MDA-MB-231, MDA-MB-468 and MCF7 cells, and autophagy inhibition impaired Huaier-induced cell death in these cancer cells. Huaier-induced cytotoxicity in human breast cancer cells was partialy due to activation of autophagy (48). In addition, Huaier augmented tamoxifen-induced autophagy in ER-positive breast cancer cells (49). We demonstrated that Huaier extract dramatically triggered autophagy in PC3 cells through electron microscopy observation, acridine orange staining, western blotting and immunofluorescence assay. Moreover, inhibition of autophagy via drugs or siRNAs significantly abrogated Huaier-induced cytotoxicity in PC3 cells. Therefore, autophagic cell death is involved in Huaier-induced cytotoxicity in human prostate cancer cells.

In this study, we demonstrated that Huaier-induced cytotoxicity and decreased cell mobility were at least partially mediated by downregulation of Lamin B1 and autophagic cell death in prostate cancer PC3 cells. The multiple mechanisms reported in our study contribute to the understanding of the complex anti-prostate cancer effects of Huaier. The present study provides a new theoretical basis for the clinical application of Huaier in prostate cancer.

Acknowledgements

Not applicable.

Funding

The present study was financially supported by the National Natural Science Foundation of China (nos. 81403147 and 81402219), the Excellent Young Scientist Foundation of Beijing University of Chinese Medicine (no. 2015-JYB-XYQ-004), and the Outstanding Young Talent Foundation of the Organization Department of Beijing Municipal Party Committee (no. 2014000021469G221).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

AY designed and performed the experiments, analyzed data and wrote the manuscript. YaZ, YW, XZ and YuZ analyzed data and revised the manuscript. PT and ZH supervised the study, designed experiments and revised the manuscript. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

TCM

Traditional Chinese Medicine

FBS

fetal bovine serum

OD

optical density

CCK-8

Cell Counting Kit-8

TEM

transmission electron microscopy

References

1 

Denmeade SR and Isaacs JT: A history of prostate cancer treatment. Nat Rev Cancer. 2:389–396. 2002. View Article : Google Scholar : PubMed/NCBI

2 

Trewartha D and Carter K: Advances in prostate cancer treatment. Nat Rev Drug Discov. 12:823–824. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Kelloff GJ: Perspectives on cancer chemoprevention research and drug development. Adv Cancer Res. 78:199–334. 2000. View Article : Google Scholar : PubMed/NCBI

4 

Hwang C: Overcoming docetaxel resistance in prostate cancer: A perspective review. Ther Adv Med Oncol. 4:329–340. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Wallace TJ, Torre T, Grob M, Yu J, Avital I, Brücher B, Stojadinovic A and Man YG: Current approaches, challenges and future directions for monitoring treatment response in prostate cancer. J Cancer. 5:3–24. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Wang CY, Bai XY and Wang CH: Traditional Chinese medicine: A treasured natural resource of anticancer drug research and development. Am J Chin Med. 42:543–559. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Xu H, Zhao X, Liu X, Xu P, Zhang K and Lin X: Antitumor effects of traditional Chinese medicine targeting the cellular apoptotic pathway. Drug Des Devel Ther. 9:2735–2744. 2015.PubMed/NCBI

8 

Yan Z, Lai Z and Lin J: Anticancer properties of traditional Chinese medicine. Comb Chem High Throughput Screen. 20:423–429. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Song X, Li Y, Zhang H and Yang Q: The anticancer effect of Huaier (Review). Oncol Rep. 34:12–21. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Hu Z, Yang A, Su G, Zhao Y, Wang Y, Chai X and Tu P: Huaier restrains proliferative and invasive potential of human hepatoma SKHEP-1 cells partially through decreased Lamin B1 and elevated NOV. Sci Rep. 6:312982016. View Article : Google Scholar : PubMed/NCBI

11 

Li Y, Qi W, Song X, Lv S, Zhang H and Yang Q: Huaier extract suppresses breast cancer via regulating tumor-associated macrophages. Sci Rep. 6:200492016. View Article : Google Scholar : PubMed/NCBI

12 

Yan X, Lyu T, Jia N, Yu Y, Hua K and Feng W: Huaier aqueous extract inhibits ovarian cancer cell motility via the AKT/GSK3β/β-catenin pathway. PLoS One. 8:e637312013. View Article : Google Scholar : PubMed/NCBI

13 

Wang X, Zhang N, Huo Q and Yang Q: Anti-angiogenic and antitumor activities of Huaier aqueous extract. Oncol Rep. 28:1167–1175. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Wu T, Chen W, Liu S, Lu H, Wang H, Kong D, Huang X, Kong Q, Ning Y and Lu Z: Huaier suppresses proliferation and induces apoptosis in human pulmonary cancer cells via upregulation of miR-26b-5p. FEBS Lett. 588:2107–2114. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Yang AL, Hu ZD and Tu PF: Research progress on anti-tumor effect of Huaier. Zhongguo Zhong Yao Za Zhi. 40:4805–4810. 2015.PubMed/NCBI

16 

Wang X, Zhang N, Huo Q, Sun M, Lv S and Yang Q: Huaier aqueous extract suppresses human breast cancer cell proliferation through inhibition of estrogen receptor α signaling. Int J Oncol. 43:321–328. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Yan L, Liu X, Yin A, Wei Y, Yang Q and Kong B: Huaier aqueous extract inhibits cervical cancer cell proliferation via JNK/p38 pathway. Int J Oncol. 47:1054–1060. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Yang A, Fan H, Zhao Y, Zha X, Zhang H, Hu Z and Tu P: Huaier aqueous extract inhibits proliferation and metastasis of tuberous sclerosis complex cell models through downregulation of JAK2/STAT3 and MAPK signaling pathways. Oncol Rep. 36:1491–1498. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Coradeghini R, Barboro P, Rubagotti A, Boccardo F, Parodi S, Carmignani G, D'Arrigo C, Patrone E and Balbi C: Differential expression of nuclear lamins in normal and cancerous prostate tissues. Oncol Rep. 15:609–613. 2006.PubMed/NCBI

20 

Klionsky DJ: Autophagy: From phenomenology to molecular understanding in less than a decade. Nat Rev Mol Cell Biol. 8:931–937. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Hu Z, Wang Y, Huang F, Chen R, Li C, Wang F, Goto J, Kwiatkowski DJ, Wdzieczak-Bakala J, Tu P, et al: Brain-expressed X-linked 2 is pivotal for hyperactive mechanistic target of rapamycin (mTOR)-mediated tumorigenesis. J Biol Chem. 290:25756–25765. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Ylä-Anttila P, Vihinen H, Jokitalo E and Eskelinen EL: Monitoring autophagy by electron microscopy in Mammalian cells. Methods Enzymol. 452:143–164. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Dittmer TA and Misteli T: The lamin protein family. Genome Biol. 12:2222011. View Article : Google Scholar : PubMed/NCBI

24 

Sun S, Xu MZ, Poon RT, Day PJ and Luk JM: Circulating Lamin B1 (LMNB1) biomarker detects early stages of liver cancer in patients. J Proteome Res. 9:70–78. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Li L, Du Y, Kong X, Li Z, Jia Z, Cui J, Gao J, Wang G and Xie K: Lamin B1 is a novel therapeutic target of betulinic acid in pancreatic cancer. Clin Cancer Res. 19:4651–4661. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Tsujimoto Y and Shimizu S: Another way to die: Autophagic programmed cell death. Cell Death Differ. 12 Suppl 2:S1528–S1534. 2005. View Article : Google Scholar

27 

Fulda S and Kögel D: Cell death by autophagy: Emerging molecular mechanisms and implications for cancer therapy. Oncogene. 34:5105–5113. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Petibone DM, Majeed W and Casciano DA: Autophagy function and its relationship to pathology, clinical applications, drug metabolism and toxicity. J Appl Toxicol. 37:23–37. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Dunn WA Jr: Studies on the mechanisms of autophagy: Formation of the autophagic vacuole. J Cell Biol. 110:1923–1933. 1990. View Article : Google Scholar : PubMed/NCBI

30 

Kim AD, Kang KA, Kim HS, Kim DH, Choi YH, Lee SJ, Kim HS and Hyun JW: A ginseng metabolite, compound K, induces autophagy and apoptosis via generation of reactive oxygen species and activation of JNK in human colon cancer cells. Cell Death Dis. 4:e7502013. View Article : Google Scholar : PubMed/NCBI

31 

Noda NN and Inagaki F: Mechanisms of autophagy. Annu Rev Biophys. 44:101–122. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Li X, Li Y, Fang S, Su J, Jiang J, Liang B, Huang J, Zhou B, Zang N, Ho W, et al: Downregulation of autophagy-related gene ATG5 and GABARAP expression by IFN-λ1 contributes to its anti-HCV activity in human hepatoma cells. Antiviral Res. 140:83–94. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Ma K, Fu W, Tang M, Zhang C, Hou T, Li R, Lu X, Wang Y, Zhou J, Li X, et al: PTK2-mediated degradation of ATG3 impedes cancer cells susceptible to DNA damage treatment. Autophagy. 13:579–591. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Tanida I, Ueno T and Kominami E: LC3 and autophagy. Methods Mol Biol. 445:77–88. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Mizushima N and Yoshimori T: How to interpret LC3 immunoblotting. Autophagy. 3:542–545. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Kang R, Zeh HJ, Lotze MT and Tang D: The Beclin 1 network regulates autophagy and apoptosis. Cell Death Differ. 18:571–580. 2011. View Article : Google Scholar : PubMed/NCBI

37 

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

38 

Valastyan S and Weinberg RA: Tumor metastasis: Molecular insights and evolving paradigms. Cell. 147:275–292. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Mehlen P and Puisieux A: Metastasis: A question of life or death. Nat Rev Cancer. 6:449–458. 2006. View Article : Google Scholar : PubMed/NCBI

40 

Bubendorf L, Schöpfer A, Wagner U, Sauter G, Moch H, Willi N, Gasser TC and Mihatsch MJ: Metastatic patterns of prostate cancer: An autopsy study of 1,589 patients. Hum Pathol. 31:578–583. 2000. View Article : Google Scholar : PubMed/NCBI

41 

Park JC and Eisenberger MA: Advances in the treatment of metastatic prostate cancer. Mayo Clin Proc. 90:1719–1733. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Harborth J, Elbashir SM, Bechert K, Tuschl T and Weber K: Identification of essential genes in cultured mammalian cells using small interfering RNAs. J Cell Sci. 114:4557–4565. 2001.PubMed/NCBI

43 

Levine B and Klionsky DJ: Development by self-digestion: Molecular mechanisms and biological functions of autophagy. Dev Cell. 6:463–477. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Gozuacik D and Kimchi A: Autophagy as a cell death and tumor suppressor mechanism. Oncogene. 23:2891–2906. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Yang WL, Perillo W, Liou D, Marambaud P and Wang P: AMPK inhibitor compound C suppresses cell proliferation by induction of apoptosis and autophagy in human colorectal cancer cells. J Surg Oncol. 106:680–688. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Yao Z, Xie F, Li M, Liang Z, Xu W, Yang J, Liu C, Li H, Zhou H and Qu LH: Oridonin induces autophagy via inhibition of glucose metabolism in p53-mutated colorectal cancer cells. Cell Death Dis. 8:e26332017. View Article : Google Scholar : PubMed/NCBI

47 

Yang ZJ, Chee CE, Huang S and Sinicrope FA: The role of autophagy in cancer: Therapeutic implications. Mol Cancer Ther. 10:1533–1541. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Wang X, Qi W, Li Y, Zhang N, Dong L, Sun M, Cun J, Zhang Y, Lv S and Yang Q: Huaier extract induces autophagic cell death by inhibiting the mTOR/S6K pathway in breast cancer cells. PLoS One. 10:e01317712015. View Article : Google Scholar : PubMed/NCBI

49 

Qi W, Sun M, Kong X, Li Y, Wang X, Lv S, Ding X, Gao S, Cun J, Cai C, et al: Huaier extract synergizes with tamoxifen to induce autophagy and apoptosis in ER-positive breast cancer cells. Oncotarget. 7:26003–26015. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2018
Volume 39 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang A, Zhao Y, Wang Y, Zha X, Zhao Y, Tu P and Hu Z: Huaier suppresses proliferative and metastatic potential of prostate cancer PC3 cells via downregulation of Lamin B1 and induction of autophagy. Oncol Rep 39: 3055-3063, 2018
APA
Yang, A., Zhao, Y., Wang, Y., Zha, X., Zhao, Y., Tu, P., & Hu, Z. (2018). Huaier suppresses proliferative and metastatic potential of prostate cancer PC3 cells via downregulation of Lamin B1 and induction of autophagy. Oncology Reports, 39, 3055-3063. https://doi.org/10.3892/or.2018.6358
MLA
Yang, A., Zhao, Y., Wang, Y., Zha, X., Zhao, Y., Tu, P., Hu, Z."Huaier suppresses proliferative and metastatic potential of prostate cancer PC3 cells via downregulation of Lamin B1 and induction of autophagy". Oncology Reports 39.6 (2018): 3055-3063.
Chicago
Yang, A., Zhao, Y., Wang, Y., Zha, X., Zhao, Y., Tu, P., Hu, Z."Huaier suppresses proliferative and metastatic potential of prostate cancer PC3 cells via downregulation of Lamin B1 and induction of autophagy". Oncology Reports 39, no. 6 (2018): 3055-3063. https://doi.org/10.3892/or.2018.6358