Alterations in enhancer of zeste homolog 2, matrix metalloproteinase-2 and tissue inhibitor of metalloproteinase-2 expression are associated with ex vivo and in vitro bone metastasis in renal cell carcinoma

  • Authors:
    • Jiang Wang
    • Ye Ren
    • Xin Guo
    • Hao Cheng
    • Yaping Ye
    • Jun Qi
    • Caihong Yang
    • Hongbo You
  • View Affiliations

  • Published online on: January 8, 2015     https://doi.org/10.3892/mmr.2015.3164
  • Pages: 3585-3592
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Renal cell carcinoma (RCC) has a high potential for bone metastasis; however, the molecular mechanisms underlying this metastasis have remained to be elucidated. The present study aimed to explore the expression levels of enhancer of zeste homolog 2 (EZH2), matrix metalloproteinase-2 (MMP2) and tissue inhibitor of metalloproteinase-2 (TIMP2) as determinants of RCC-associated bone metastasis. Their expression was evaluated in a newly generated RCC cell subline that has a high potential for bone metastasis, in tissue specimens from metastasized bone tissues from patients with RCC and in RCC tissues without metastasis. A total of 25 RCC tissue specimens without metastasis and 13 RCC tissue specimens with bone metastasis were acquired for immunohistochemical analysis of EZH2, MMP2 and TIMP2 protein expression. The expression levels of EZH2, MMP2 and TIMP2 mRNA and protein were analyzed in the ACHN and ACHN-BO5 cell lines using western blot and reverse transcription polymerase chain reaction (PCR) analyses. Methylation-specific PCR was also used to analyze TIMP2 promoter methylation. EZH2 and MMP2 proteins were found to be expressed at higher levels in tissues from patients where RCC had metastasized to the bone as compared with those in RCC patients without metastasis, whereas there was no significant difference in the expression of TIMP2 protein between the two tissues. Furthermore, the expression of EZH2 protein was correlated with MMP2 expression, but there was no significant correlation between the expression of EZH2 and TIMP2 proteins. The in vitro results using cell lines confirmed the ex vivo findings, indicating that the expression levels of EZH2 and MMP2 protein and mRNA were higher in ACHN-BO5 cells than those in ACHN cells. By contrast, TIMP2 protein and mRNA expression levels were lower in ACHN-BO5 cells than those in the parental ACHN cells. The TIMP2 promoter was highly methylated in ACHN-BO5 cells compared with that in ACHN cells. Upregulation of EZH2, MMP2 and TIMP2 expression was correlated with metastasis of RCC to bone tissues ex vivo and in vitro. Further studies are required in order to elucidate the mechanism underlying the altered expression of these genes.

Introduction

Renal cell carcinoma (RCC), which has been suggested to originate from the renal tubules and collecting tube epithelial cells, accounts for 85% of malignant kidney neoplasms and ~2% of all human malignancies (1,2). RCC is a pathologically heterogeneous disease, which can be classified into clear, papillary, granular, spindle and mixed cell subtypes based on certain cytoplasmic features (3). RCC morbidity increases by 2% annually and mortality has reached ~100,000 cases/year worldwide (4). Approximately 30% of patients with RCC develop metastatic disease, most frequently in the lungs, bones or brain (5). A clinical study confirmed that osteolysis represented 30% of the total metastatic disease cases associated with RCC (6). The incidence rate of bone tissue metastasis was higher in autopsy data from patients with RCC (5). The prognosis of RCC patients is influenced by a variety of factors, including tumor size, invasion, metastasis, histological type and nuclear grade (7); the five-year survival rate of patients with RCC was 90% for stage I, 51% for stage II, 22% for stage III and 4.6% for stage IV (8). Therefore, the elucidation of the key factors and molecular mechanisms underlying the metastasis of RCC to bone is required. A previous study by our group established an ACHN cell subline (ACHN-BO5) with high potential of bone metastasis compared with that of ACHN cells in animals in order to aid the elucidation of the underlying molecular mechanisms (9). In this subline, the pathological karyokinesis was significantly increased, which indicated that the malignant phenotype of the ACHN subline was higher than that of the parental ACHN cells. Following five passages of in vitro culture, the subline was named ‘ACHN-BO5’. Subsequently, the gene alterations responsible for the high potential of bone metastasis were investigated using a complementary DNA (cDNA) microarray analysis to compare ACHN-BO5 cells with the parental ACHN cells. Alterations in the expression of enhancer of zeste homolog 2 (EZH2) and matrix metalloproteinase-2 (MMP2) were detected in ACHN-BO5 cells. EZH2 is involved in maintaining the transcriptional repressive state in cells and mutation of EZH2 causes Weaver syndrome (10), a congenital disorder associated with rapid growth beginning in the prenatal period, resulting in a characteristic facial appearance and certain skeletal features (11). In addition, studies have demonstrated that altered EZH2 expression promotes human cancer development (1216). Proteins of the MMP family degrade or break down the extracellular matrix during normal physiological processes, including embryonic development and tissue remodeling, but also have a significant role in tumor metastasis (17). By contrast, tissue inhibitor of metalloproteinase-2 (TIMP2) is a natural matrix metalloproteinase inhibitor. Therefore, these proteins may have a role in mediating the metastasis of RCC to bone. In the present study, the expression of EZH2, MMP2 and TIMP2 were evaluated in RCC tissue specimens with or without bone metastasis and in ACHN-BO5 and ACHN cells to elucidate the correlation between their expression and metastasis in bone. Methylation-specific PCR analysis of TIMP2 promoter methylation was also performed. The results of the present study may aid the elucidation of the mechanisms underlying RCC metastasis and provide potential therapeutic targets for the prevention or treatment of RCC metastasis.

Materials and methods

Cell lines and culture

The human renal carcinoma cell line ACHN was obtained from the China Center of Type Culture Collection (Wuhan, China). The ACHN-B05 cell line was a subline of ACHN with high potential of bone metastasis, established in a previous study by our group (9). These cell lines were cultured in Dulbecco’s modified Eagle’s medium (DMEM; Invitrogen Life Technologies, Carlsbad, CA, USA) supplemented with 10% fetal bovine serum (FBS; Invitrogen Life Technologies) at 37°C in a humidified incubator with 5% CO2.

Tissue specimens and immunohistochemistry

Primary renal cancer biopsy specimens (n=25) and biopsies of renal cancer that had metastasized to bone tissues (n=13) were obtained from The Tongji Hospital affiliated with Huazhong University of Science and Technology (Wuhan, China) between March 2010 and April 2013. The normal control renal biopsy specimens were obtained from adjacent normal tissues. The present study was approved by the ethics committee of Tongji Hospital affiliated with Huazhong University of Science and Technology. Written informed consent was obtained from all patients or their family. All tissues were fixed in 4% paraformaldehyde solution (Boster Biological Technology, Ltd., Wuhan, China) for 20 min at room temperature and embedded into paraffin using a routine tissue process (18). Tissue sections (4-μM thick) were prepared from the paraffin blocks and mounted onto glass slides. For immunohistochemical analysis, tissue sections were deparaffinized and rehydrated in water. The sections were heated in a pressure cooker (121°C, 4 min) in a citric acid buffer (Boster Biological Technology, Ltd.) for antigen retrieval and then incubated with 3% H2O2/phosphate-buffered saline (PBS; Boster Biological Technology, Ltd.) at room temperature for 30 min to block potential peroxidase activity. Following incubation with 20% normal serum (Boster Biological Technology, Ltd.) for 30 min, the sections were further incubated with primary antibodies: Mouse monoclonal immunoglobulin G1 (IgG1) anti-MMP2 antibody at a dilution of 1:800 (sc-13594; Santa Cruz Biotechnology, Inc., Dallas, TX, USA), goat polyclonal IgG anti-TIMP2 antibody at a dilution of 1:600 (sc-6835; Santa Cruz Biotechnology Inc.) or an goat polyclonal IgG anti-EZH2 antibody at a dilution of 1:100 (E6906; Sigma-Aldrich, St. Louis, MO, USA) overnight at 4°C. The following day, the sections were washed three times with PBS and subsequently incubated with the secondary antibodies (anti-mouse IgG and anti-goat IgG horseradish peroxidase; Boster Biological Technology, Ltd.) for 1 h at 37°C. A color reaction was performed using 3,3′-diaminobenzidine (Boster Biological Technology, Ltd.) as the chromogen. Diluted Sav-HRP conjugates were applied to the sections on the slides and incubated in a humidified chamber at room temperature for 30 min (protected from the light). Slides were washed with PBS twice, for 5 min each. DAB substrate solution (freshly made just before use: 0.05% DAB - 0.015% H2O2 in PBS) was applied to the sections on the slides to reveal the color of antibody staining. The stained tissue sections were independently reviewed and scored under an Olympus CKX31 inverted microscope (Olympus Corp., Tokyo, Japan) by two investigators. The statistical results were analyzed using Sigmaplot 11.0 software (Systat Software, Inc., Chicago, IL, USA).

DNA extraction and methylation-specific PCR (MSP)

Genomic DNA was extracted from ACHN and ACHN-BO5 cell lines using a Genomic DNA extraction kit (Boehringer, Mannheim, Germany) according to the manufacturer’s instructions. DNA concentration, purity and integrity were measured using a spectrophotometer (Gilford 250; Gilford Instrument Laboratories, Inc., Oberlin, OH, USA) and gel electrophoresis. For MSP, genomic DNA samples (2 μg) were denatured with sodium hydroxide chemically modified with sodium bisulfite (Boster Biological Technology, Ltd.). An MSP primer for the amplification of the TIMP2 promoter was designed using Methprime software (http://www.urogene.org/methprimer/index1.html). The methylation primers of the TIMP2 promoter were as follows: Forward, 5′-TTTTATTGTAGGAAAGGTCGA-3′ and reverse, 5′-GAAATCATAAAACAACGCGTA-3′, which amplified a 159-bp PCR product. The demethylation primers of the TIMP2 promoter used were: Forward, 5′-GAAGGAATATTTTATTGTAGGAAAGGTT-3′ and reverse, 5′-TATAACACAAAATCATAAAACAACACATA-3′, which amplified a 176-bp PCR product. PCR amplification occurred in a final volume of 25 μl containing: 2.5 μl PCR buffer, 2 μl MgCl2, 2.5 μl deoxynucleotide triphosphate mixture, 1 μl of each primer, 5 μl modified DNA template, 10.85 μl sterilized deionized water and 0.15 μl Taq enzyme (Takara Bio, Inc., Otsu, Japan). The PCR conditions were set at a pre-denaturation temperature of 94°C for 5 min followed by 45 cycles of 94°C for 30 sec, 60°C (methylated) or 55°C (demethylated) for 45 sec, 75°C for 40 sec and a final extension at 73°C for 5 min. Genomic DNA chemically modified with sss1 methylation enzyme (New England Biolabs, Shanghai, China) and bisulfite salts (Sigma-Aldrich) was used as a positive control. Deionized water was used as a negative control. PCR products were isolated using agarose gel electrophoresis and imaged under an ultraviolet lamp (XX15B, Spectronics Corp., Westbury, NY, USA).

RNA isolation and semi-quantitative RT-PCR

The expression levels of EZH2, MMP2 and TIMP2 mRNA were evaluated using RT-PCR analysis. Briefly, total cellular RNA was isolated from cells using TRIzol reagent (Invitrogen Life Technologies) and reverse-transcribed to cDNA using the First Strand cDNA Synthesis kit (Rever Tra Ace-α; ToYoBo Co., Ltd., Osaka, Japan) according to the manufacturer’s instructions. The primer sequences of genes and fragment sizes are indicated in Table I. The PCR conditions were as follows: Pre-denaturation at 94°C for 5 min and 32 cycles of 94°C for 45 sec; 52.5°C, 56°C and 54°C as the annealing temperature for 1 min, 72°C for 90 sec and a final extension at 72°C for 10 min. The PCR product was subsequently sequenced and identified by gel electrophoresis.

Table I

Primers for PCR amplification of gene expression.

Table I

Primers for PCR amplification of gene expression.

GeneSequencesSize of PCR products (bp)
EZH2 5′-GTGGAGAGATTATTTCTCAAGATG-3′
5′-CCGACATACTTCAGGGCATCAGCC-3′289
MMP2 5′-GAGAACCAAAGTCTGAAGAG-3′
5′-GGAGTGAGAATGCTGATTAG-3′207
TIMP25′-CCTCGGCCTTTC CTGCAAT-3′
5′-TATCTACACGGCCCCCTCCT-3′89
GAPDH 5′-GAAGGTGAAGGTCGGAGTC-3′
5′-GAAGATGGTGATGGGATTTC-3′226

[i] PCR, polymerase chain reaction; EZH2, enhancer of zeste homolog 2; MMP2, matrix metalloproteinase-2; TIMP2, tissue inhibitor of metalloproteinase-2.

Protein extraction and western blot analysis

Cells were collected and lysed with a pre-cooled cell lysis buffer (Boster Biological Technology, Ltd.) containing 100 mM Tris-HCl, 500 mM EDTA, 20 mM NaCl, and 10% SDS. The protein concentration was determined using a Bicinchoninic Acid Assay kit (Sigma-Aldrich, Irvine, Scotland). Briefly, an equal amount of protein sample was separated by SDS-PAGE and transferred onto a polyvinylidene fluoride membrane (A-FIT Biosciences, Beijing, China). For western blot analysis, the membranes were incubated in 5% skimmed milk/PBS at room temperature for 1 h and then further incubated with anti-MMP2 antibody at a dilution of 1:800, anti-TIMP2 antibody at a dilution of 1:600 or anti-EZH2 antibody at a dilution of 1:100 at 4°C overnight. The following day, the membranes were washed three times with PBS-Tween-20 and incubated with a horseradish peroxidase-conjugated secondary antibody at a dilution of 1:7,500 at room temperature for 2 h. Immunoreactive proteins were visualized using enhanced chemiluminescence (Pierce Biotechnology, Inc., Rockford, IL, USA) according to the manufacturer’s instructions and exposed to x-ray films (Kodak, Rochester, NY, USA). The expression levels of these proteins were normalized to an internal control, GAPDH.

Statistical analysis

Values are presented as the mean ± standard deviation of three independent experiments. All statistical analyses were performed using SPSS 11.0 software (SPSS, Inc., Chicago, IL, USA). The expression levels of EZH2, MMP2, TIMP2 mRNA and protein in ACHN and ACHN-B05 cells were compared using one-way analysis of variance. Differences in TIMP2 promoter methylation were analyzed using a χ2 test. A Pearson’s correlation test was used to analyze the associations between different groups. P<0.05 was considered to indicate a statistically significant difference between values.

Results

Differential expression of EZH2, MMP2 and TIMP2 proteins in tissues of patients with RCC as well as ACHN and ACHN-B05 cells

In the present study, the expression of EZH2, MMP2 and TIMP2 proteins in tissues from patients with RCC with and without bone metastasis were evaluated by immunohistochemical analysis. The expression of EZH2 protein was higher in tissues from patients with RCC that had metastasized to the bone than in tissues of patients with RCC without metastasis (P=0.031; Fig. 1A–D). Analogously, expression levels of MMP2 protein were also higher in tissues from patients where RCC had metastatized than those in patients with RCC without metastasis (P=0.047; Fig. 2A–D). By contrast, there were no significant differences in the expression of TIMP2 protein between the tissue types (P=0.2932; Fig. 3A–D). Furthermore, the expression of EZH2 and MMP2 proteins were found to be correlated (r=0.6652; P=0.0131; Fig. 4A), whereas there was no significant correlation between EZH2 and TIMP2 protein expression (r=−0.5484; P=0.0523; Fig. 4B).

Western blot analysis was used to investigate the expression levels of EZH2 protein in ACHN and ACHN-BO5 cells. EZH2 protein expression levels were demonstrated to be higher in ACHN-BO5 cells, a sub-line of ACHN with a higher potential for metastasis to the bone, than those in the parental ACHN cells (P<0.05), suggesting that EZH2 protein may be involved in mediating the metastasis of RCC to bone (Fig. 5A). In addition, the expression levels of MMP2 protein were higher in ACHN-BO5 cells than those in the parental ACHN cells (P<0.05). By contrast, TIMP2 protein expression levels were lower in ACHN-BO5 cells than those in the parental ACHN cells (P<0.05; Fig. 5B).

EZH2, MMP2 and TIMP2 mRNA expression levels differ between ACHN and ACHN-B05 cells

RT-PCR analysis was performed in order to evaluate whether the altered expression of these three proteins occurred at the transcriptional level in the ACHN and ACHN-BO5 cell lines. The expression levels of EZH2 and MMP2 mRNA were higher in ACHN-BO5 cells than those in ACHN cells, whereas the expression levels of TIMP2 mRNA were lower in ACHN-BO5 cells than those in ACHN cells (Fig. 6).

TIMP2 is more highly methylated in ACHN-BO5 cells

The potential mechanism underlying the downregulated expression of TIMP2 mRNA in ACHN-BO5 cells was assessed using MSP. The TIMP2 promoter was more highly methylated in ACHN-BO5 cells than in ACHN cells (Fig. 7).

Discussion

Cancer metastasis is a complex process, where cancer cells migrate from their site of origin and invade other parts of the body via the bloodstream, lymphatic system or direct extension (19). Molecularly, tumor cells gain gene transcription capabilities and express various proteins and enzymes in order to degrade extracellular matrix proteins and invade adjacent tissues (20). However, the distant metastasis of various types of human cancer indicates preferences for certain organs; therefore, certain types of cancer tend to spread to particular organs and tissues (19). For example, breast cancer preferentially metastasizes to bone and lung tissue (21). RCC frequently metastasizes to the lungs, bone or brain, whereas the brain is most commonly the distant site of metastasis of melanoma (22). Therefore, a study of organ-specific RCC metastasis may aid in the prevention of RCC progression. In a previous study by our group, an RCC cell line (ACHN-BO5) with high potential for metastasis to the bone was generated (9). These cells exhibited a significantly enhanced invasion and proliferation capacity in vitro, compared to that of the parental ACHN cells. The present study investigated whether cell adhesion molecules, including EZH2, MMP2 and TIMP2, contributed to alterations in the phenotype of tumor cells. Expression levels of EZH2 and MMP2 mRNA and protein were higher in ACHN-BO5 cells than those in ACHN cells, whereas the expression of TIMP2 mRNA and protein was lower in ACHN-BO5 cells than that in ACHN cells. MSP data indicated that the downregulated expression of TIMP2 may be due to the methylation of the TIMP2 promoter. Furthermore, the expression of these proteins was evaluated in tissue specimens from patients with RCC that had metastasized to the bone and patients with RCC without metastasis. The results confirmed those of the in vitro investigations, indicating that the expression of EZH2 and MMP2 protein was higher in tissues from patients with RCC that had metastasized to the bone than that in tissues from renal cancer patients without metastasis; and that there was no significant difference in the expression of TIMP2 protein between the two types of tissue.

The epigenetic modification enzyme, EZH2, has a homologous structure to the Drosophila E(z) gene, and was identified in the 1990s (23). Cardoso et al (24) demonstrated that the EZH2 protein was a key member of the polycomb group gene family and was able to modulate cell proliferation and the signaling pathway via the suvar3–9, enhancer of zeste, trtharax domain. The EZH2 protein is involved in mediating the development, metastasis, invasiveness and prognosis of various types of human cancer (2332). Therefore, EZH2 is of interest in basic and clinical studies of cancer. Several studies have demonstrated that EZH2 protein is highly expressed in RCC tissues (3336), the level of which is associated with RCC dedifferentiation, suggesting that the EZH2 protein may contribute to RCC progression and metastasis. More recently, studies have indicated that EZH2 protein is highly expressed in prostate cancer tissues, which are also associated with a high rate of metastasis to the bone (16,37,38). Knockdown of EZH2 expression in metastatic bone tumors leads to atrophy of the metastatic bone foci and a reduction in or end to bone destruction (39), suggesting that EZH2 protein has a role in mediating metastasis to the bone.

MMPs, including MMP2, are required for degradation of the extracellular matrix and are specifically inhibited by the TIMPs. Therefore, these two families of proteins have significant roles in tumor metastasis. Numerous studies have confirmed that alterations in the balance of MMPs/TIMPs may lead to the metastasis of human cancers to the bone (4043). Further studies have demonstrated that a downregulation of TIMP2 expression was associated with methylation of the TIMP2 promoter (4450). Of note, EZH2 contains histone methyltransferase activity, which is able to silence genes via the methylation of H3 histone lysine 27 (51). It was therefore hypothesized that overexpression of EZH2 protein may lead to epigenetic silencing of TIMP2 expression. The results of the present study confirmed that there was an enhanced level of TIMP2 promoter methylation in ACHN-BO5 cells compared to that in ACHN cells. However, the results of the ex vivo investigations did not demonstrate an association between EZH2 expression and reduced TIMP2 protein, which does not support the hypothesis that EZH2 protein may be able to cause epigenetic silencing of TIMP2 expression. Due to the small sample size in the present study, further investigations are required in order to confirm this hypothesis.

Further studies are required to elucidate the molecular mechanisms underlying how the expression of EZH2, MMP2 and TIMP2 is altered in RCC tissues or cells with high potential for bone metastasis.

Acknowledgements

The present study was part of the programs on The Natural Science Foundation funded through The Science and Technology Department of Hubei Province, Wuhan, China (no. 2008CDB168) and The Science Foundation funded through The Health Department of Hubei Province, Wuhan, China (no. 2010JX5B03). This study was also part of the programs on The Nature Science Foundation funded by The Huazhong University of Science and Technology (2013QN200). The authors would like to thank all study participants and Medjaden Bioscience Limited (Hong Kong, China) for assisting in the preparation of this manuscript.

References

1 

Kosary C and McLaughlin J: Kidney and renal pelvis. Cancer Statistics Review: 1973–1990. Miller BA, Ries LAG, Hankey BE, et al: National Institutes Of Health; Bethesda: pp. x1–x22. 1993

2 

Fleming S and Lewi H: Collecting duct carcinoma of the kidney. Histopathology. 10:1131–1141. 1986. View Article : Google Scholar : PubMed/NCBI

3 

Thoenes W, Störkel S and Rumpelt H: Histopathology and classification of renal cell tumors (adenomas, oncocytomas and carcinomas). The basic cytological and histopathological elements and their use for diagnostics. Pathol Res Pract. 181:125–143. 1986. View Article : Google Scholar : PubMed/NCBI

4 

Pisani P, Parkin D and Ferlay J: Estimates of the worldwide mortality from eighteen major cancers in 1985. Implications for prevention and projections of future burden. Int J Cancer. 55:891–903. 1993. View Article : Google Scholar : PubMed/NCBI

5 

Ritchie A and Chisholm G: The natural history of renal carcinoma. Semin Oncol. 10:390–400. 1983.PubMed/NCBI

6 

Wood SL and Brown JE: Skeletal metastasis in renal cell carcinoma: current and future management options. Cancer Treat Rev. 38:284–291. 2012. View Article : Google Scholar

7 

Rini BI, Campbell SC and Escudier B: Renal cell carcinoma. Lancet. 373:1119–1132. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Erdoğan F, Demirel A and Polat O: Prognostic significance of morphologic parameters in renal cell carcinoma. Int J Clin Pract. 58:333–336. 2004. View Article : Google Scholar

9 

Wang J, Chen A, Yang C, Zeng H, Qi J and Guo FJ: A bone-seeking clone exhibits different biological properties from the ACHN parental human renal cell carcinoma in vivo and in vitro. Oncol Rep. 27:1104–1110. 2012.

10 

Gibson WT, Hood RL, Zhan SH, et al; FORGE Canada Consortium. Mutations in EZH2 cause Weaver syndrome. Am J Hum Genet. 90:110–118. 2012. View Article : Google Scholar :

11 

Weaver DD, Graham CB, Thomas I and Smith DW: A new overgrowth syndrome with accelerated skeletal maturation, unusual facies, and camptodactyly. J Pediatr. 84:547–552. 1974. View Article : Google Scholar : PubMed/NCBI

12 

Kleer CG, Cao Q, Varambally S, et al: EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proc Natl Acad Sci USA. 100:11606–11611. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Kehinde EO, Maghrebi M and Anim JT: The importance of determining the aggressiveness of prostate cancer using serum and tissue molecular markers. Can J Urol. 15:3967–3974. 2008.PubMed/NCBI

14 

Simon JA and Lange CA: Roles of the EZH2 histone methyltransferase in cancer epigenetics. Mutat Res. 647:21–29. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Varambally S, Cao Q, Mani RS, et al: Genomic loss of microRNA-101 leads to overexpression of histone methyltransferase EZH2 in cancer. Science. 322:1695–1699. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Shen L, Cui J, Liang S, Pang Y and Liu P: Update of research on the role of EZH2 in cancer progression. Onco Targets Ther. 6:321–324. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Nagase H and Woessner JF Jr: Matrix metalloproteinases. J Biol Chem. 274:21491–21494. 1999. View Article : Google Scholar : PubMed/NCBI

18 

Shi SR, Key ME and Kalra KL: Antigen retrieval in formalin-fixed, paraffin-embedded tissues: an enhancement method for immunohistochemical staining based on microwave oven heating of tissue sections. J Histochem Cytochem. 39:741–748. 1991. View Article : Google Scholar : PubMed/NCBI

19 

Nguyen DX and Massagué J: Genetic determinants of cancer metastasis. Nat Rev Genet. 8:341–352. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Wong GS and Rustgi AK: Matricellular proteins: priming the tumour microenvironment for cancer development and metastasis. Br J Cancer. 108:755–761. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Drabsch Y and ten Dijke P: TGF-β signaling in breast cancer cell invasion and bone metastasis. J Mammary Gland Biol Neoplasia. 16:97–108. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Zlotnik A, Burkhardt AM and Homey B: Homeostatic chemokine receptors and organ-specific metastasis. Nat Rev Immunol. 11:597–606. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Chen H, Rossier C and Antonarakis SE: Cloning of a human homolog of the Drosophila enhancer of zeste gene (EZH2) that maps to chromosome 21q22.2. Genomics. 38:30–37. 1996. View Article : Google Scholar : PubMed/NCBI

24 

Cardoso C, Timsit S, Villard L, Khrestchatisky M, Fontès M and Colleaux L: Specific interaction between the XNP/ATR-X gene product and the SET domain of the human EZH2 protein. Hum Mol Genet. 7:679–684. 1998. View Article : Google Scholar : PubMed/NCBI

25 

Heinen A, Tzekova N, Graffmann N, et al: Histone methyltransferase enhancer of zeste homolog 2 regulates Schwann cell differentiation. Glia. 60:1696–1708. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Xia H, Yu CH, Zhang Y, et al: EZH2 silencing with RNAi enhances irradiation-induced inhibition of human lung cancer growth in vitro and in vivo. Oncol Lett. 4:135–140. 2012.PubMed/NCBI

27 

Cho HM, Jeon HS, Lee SY, et al: microRNA-101 inhibits lung cancer invasion through the regulation of enhancer of zeste homolog 2. Exp Ther Med. 2:963–967. 2011.

28 

Kikuchi J, Takashina T, Kinoshita I, et al: Epigenetic therapy with 3-deazaneplanocin A, an inhibitor of the histone methyltransferase EZH2, inhibits growth of non-small cell lung cancer cells. Lung Cancer. 78:138–143. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Marchesi I, Fiorentino FP, Rizzolio F, Giordano A and Bagella L: The ablation of EZH2 uncovers its crucial role in rhabdomyosarcoma formation. Cell Cycle. 11:3828–3836. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Zhou J, Roh JW, Bandyopadhyay S, et al: Overexpression of enhancer of zeste homolog 2 (EZH2) and focal adhesion kinase (FAK) in high grade endometrial carcinoma. Gynecol Oncol. 128:344–348. 2013. View Article : Google Scholar

31 

Wan L, Li X, Shen H and Bai X: Quantitative analysis of EZH2 expression and its correlations with lung cancer patients’ clinical pathological characteristics. Clin Transl Oncol. 15:132–138. 2013. View Article : Google Scholar

32 

Zhang R, Wang R, Chang H, et al: Downregulation of Ezh2 expression by RNA interference induces cell cycle arrest in the G0/G1 phase and apoptosis in U87 human glioma cells. Oncol Rep. 28:2278–2284. 2012.PubMed/NCBI

33 

Wagener N, Macher-Goeppinger S, Pritsch M, et al: Enhancer of zeste homolog 2 (EZH2) expression is an independent prognostic factor in renal cell carcinoma. BMC Cancer. 10:5242010. View Article : Google Scholar : PubMed/NCBI

34 

Wagener N, Holland D, Bulkescher J, et al: The enhancer of zeste homolog 2 gene contributes to cell proliferation and apoptosis resistance in renal cell carcinoma cells. Int J Cancer. 123:1545–1550. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Hinz S, Weikert S, Magheli A, et al: Expression profile of the polycomb group protein enhancer of Zeste homologue 2 and its prognostic relevance in renal cell carcinoma. J Urol. 182:2920–2925. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Sakurai T, Bilim VN, Ugolkov AV, et al: The enhancer of zeste homolog 2 (EZH2), a potential therapeutic target, is regulated by miR-101 in renal cancer cells. Biochem Biophys Res Commun. 422:607–614. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Li K, Chen MK, Situ J, et al: Role of co-expression of c-Myc, EZH2 and p27 in prognosis of prostate cancer patients after surgery. Chin Med J (Engl). 126:82–87. 2013.

38 

Yang YA and Yu J: EZH2, an epigenetic driver of prostate cancer. Protein Cell. 4:331–341. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Takeshita F, Minakuchi Y, Nagahara S, et al: Efficient delivery of small interfering RNA to bone-metastatic tumors by using atelocollagen in vivo. Proc Natl Acad Sci USA. 102:12177–12182. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Voorzanger-Rousselot N, Juillet F, Mareau E, Zimmermann J, Kalebic T and Garnero P: Association of 12 serum biochemical markers of angiogenesis, tumour invasion and bone turnover with bone metastases from breast cancer: a crossectional and longitudinal evaluation. Br J Cancer. 95:506–514. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Andela VB, Gordon AH, Zotalis G, et al: NFkappaB: a pivotal transcription factor in prostate cancer metastasis to bone. Clin Orthop Relat Res. (415 Suppl): S75–S85. 2003. View Article : Google Scholar : PubMed/NCBI

42 

Lhoták S, Elavathil LJ, Vukmirović-Popović S, Duivenvoorden WC, Tozer RG and Singh G: Immunolocalization of matrix metalloproteinases and their inhibitors in clinical specimens of bone metastasis from breast carcinoma. Clin Exp Metastasis. 18:463–470. 2000. View Article : Google Scholar

43 

Yoneda T, Sasaki A, Dunstan C, et al: Inhibition of osteolytic bone metastasis of breast cancer by combined treatment with the bisphosphonate ibandronate and tissue inhibitor of the matrix metalloproteinase-2. J Clin Invest. 99:2509–2517. 1997. View Article : Google Scholar : PubMed/NCBI

44 

Hsu CH, Peng KL, Kang ML, et al: TET1 suppresses cancer invasion by activating the tissue inhibitors of metalloproteinases. Cell Rep. 2:568–579. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Shin YJ and Kim JH: The role of EZH2 in the regulation of the activity of matrix metalloproteinases in prostate cancer cells. PloS One. 7:e303932012. View Article : Google Scholar : PubMed/NCBI

46 

Fornari F, Milazzo M, Chieco P, et al: In hepatocellular carcinoma miR-519d is upregulated by p53 and DNA hypomethylation and targets CDKN1A/p21, PTEN, AKT3 and TIMP2. J Pathol. 227:275–285. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Matsusaka K, Kaneda A, Nagae G, et al: Classification of Epstein-Barr virus-positive gastric cancers by definition of DNA methylation epigenotypes. Cancer Res. 71:7187–7197. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Hervouet E, Vallette FM and Cartron PF: Dnmt3/transcription factor interactions as crucial players in targeted DNA methylation. Epigenetics. 4:487–499. 2009. View Article : Google Scholar : PubMed/NCBI

49 

Martinez R, Schackert G and Esteller M: Hypermethylation of the proapoptotic gene TMS1/ASC: prognostic importance in glioblastoma multiforme. J Neurooncol. 82:133–139. 2007. View Article : Google Scholar

50 

Chu LC, Eberhart CG, Grossman SA and Herman JG: Epigenetic silencing of multiple genes in primary CNS lymphoma. Int J Cancer. 119:2487–2491. 2006. View Article : Google Scholar : PubMed/NCBI

51 

Crea F, Fornaro L, Bocci G, et al: EZH2 inhibition: targeting the crossroad of tumor invasion and angiogenesis. Cancer Metastasis Rev. 31:753–761. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2015
Volume 11 Issue 5

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
Wang J, Ren Y, Guo X, Cheng H, Ye Y, Qi J, Yang C and You H: Alterations in enhancer of zeste homolog 2, matrix metalloproteinase-2 and tissue inhibitor of metalloproteinase-2 expression are associated with ex vivo and in vitro bone metastasis in renal cell carcinoma. Mol Med Rep 11: 3585-3592, 2015
APA
Wang, J., Ren, Y., Guo, X., Cheng, H., Ye, Y., Qi, J. ... You, H. (2015). Alterations in enhancer of zeste homolog 2, matrix metalloproteinase-2 and tissue inhibitor of metalloproteinase-2 expression are associated with ex vivo and in vitro bone metastasis in renal cell carcinoma. Molecular Medicine Reports, 11, 3585-3592. https://doi.org/10.3892/mmr.2015.3164
MLA
Wang, J., Ren, Y., Guo, X., Cheng, H., Ye, Y., Qi, J., Yang, C., You, H."Alterations in enhancer of zeste homolog 2, matrix metalloproteinase-2 and tissue inhibitor of metalloproteinase-2 expression are associated with ex vivo and in vitro bone metastasis in renal cell carcinoma". Molecular Medicine Reports 11.5 (2015): 3585-3592.
Chicago
Wang, J., Ren, Y., Guo, X., Cheng, H., Ye, Y., Qi, J., Yang, C., You, H."Alterations in enhancer of zeste homolog 2, matrix metalloproteinase-2 and tissue inhibitor of metalloproteinase-2 expression are associated with ex vivo and in vitro bone metastasis in renal cell carcinoma". Molecular Medicine Reports 11, no. 5 (2015): 3585-3592. https://doi.org/10.3892/mmr.2015.3164