SET and MYND domain-containing protein 3 is overexpressed in human glioma and contributes to tumorigenicity

  • Authors:
    • Bin Dai
    • Weiqing Wan
    • Peng Zhang
    • Yisong Zhang
    • Changcun Pan
    • Guolu Meng
    • Xinru Xiao
    • Zhen Wu
    • Wang Jia
    • Junting Zhang
    • Liwei Zhang
  • View Affiliations

  • Published online on: September 1, 2015     https://doi.org/10.3892/or.2015.4239
  • Pages: 2722-2730
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

SET and MYND domain-containing protein 3 (SMYD3) is a histone H3 lysine 4 (H3K4) di- and tri-methyltransferase that forms a transcriptional complex with RNA polymerase II and plays an important role in early embryonic lineage commitment through the activation of lineage-specific genes. SMYD3 activates the transcription of oncogenes and cell cycle genes in gastric and breast cancer cells. However, the contribution of SMYD3 in glioma tumorigenesis remains unknown. Here, we determined the expression of SMYD3 and assessed its clinical significance in human glioma. We found that SMYD3 was overexpressed in human glioma but not in normal brain tissue. The level of SMYD3 protein expression in human glioma tissues was directly correlated with the glioma grade. The level of SMYD3 protein expression in human glioma tissues was inversely correlated with patient survival. Enforced SMYD3 expression promoted glioma LN-18 cell proliferation. Inhibition of SMYD3 expression in glioma T98G cells suppressed their anchorage‑independent growth in vitro and tumorigenicity in vivo. Furthermore, we found that SMYD3 regulated the expression of p53 protein, which is essential in SMYD3‑induced cell growth in glioma cells. These results showed that SMYD3 is overexpressed in human glioma and contributes to glioma tumorigenicity through p53. Therefore, SMYD3 may be a new potential therapeutic target for human malignant glioma.

Introduction

Malignant gliomas are the most common primary intrinsic brain tumors and are highly lethal (1,2). Despite advances in neurosurgery, radiotherapy, and chemotherapy, the prognosis of malignant glioma remains dismal, with an estimated median survival of <1 year (1,3). With the emergence of biologically target-based therapies, it is important to identify new therapeutic targets and to develop novel targeted treatments to battle this deadly disease (1,4). Insights into the pathogenesis of gliomas are likely to arise from the study of inherited cancer syndromes, in which affected individuals are prone to the development of glial malignancies.

Epigenetics, which is defined as heritable changes in gene expression that are not coded in the DNA sequence itself, is increasingly linked with tumorigenesis (5). Among the epigenetic regulatory mechanisms, histone methylation has demonstrated the power of modifications over the DNA gene (6). A recent study identified the SET and MYND domain-containing protein 3 (SMYD3), which possesses histone methyltransferase activity responsible for catalyzing the methylation of histone H3 at K4 (7). SMYD3 interacts with its binding motif 5′-CCCTCC-3′ in the promoter region of its target genes (7). These downstream genes include several oncogenes as well as genes involved in the control of the cell cycle and signal transduction (8). The functions of such downstream genes are involved in numerous aspects of the process of cell growth and apoptosis (8). SMYD3 expression was shown to be enhanced in hepatocellular carcinoma (HCC) and involved in the growth of HCC cells (9). Overexpression of SMYD3 has been observed in breast and gastric cancer tissues (10,11). In addition, it was confirmed that SMYD3 was essential for the proliferation and migration in NIH3T3 cells (12). These results suggest that SMYD3 plays a role in the oncogenesis of certain malignancies. However, whether SMYD3 expression contributes to glioma development and progression is not known.

The p53 protein has been demonstrated to have an essential role in glioma cell proliferation, motility and invasion (13). Lower expression of p53 has been observed in glioma tissues compared with that in normal samples, and lower levels of p53 are associated with both an increased risk of lymph node metastases and poor prognoses (14). Chuikov et al demonstrated that SETD7 methylates lysine 372 of p53 (p53K372) and that this methylation enhances p53 stability and transcriptional activity. K372-methylated p53 is restricted to the nucleus, although p53 is equally distributed between the nuclear and cytosolic fractions (15). Subsequently, Huang et al showed that the PKMT SET and MYND-domain containing 2 (SMYD2) monomethylates lysine 370 of p53 (p53K370); this lysine residue is located in the regulatory domain at the extreme carboxyl terminus of p53 (16). Knockdown of SMYD2 by small interfering RNA enhances p53-mediated apoptosis in cancer cells (16). In addition, SMYD2-dependent p53K370 methylation impairs the expression of CDKN1A, an important downstream target of p53, implying that SMYD2 represses the function of p53 through K370 monomethylation. Moreover, p53K372 methylation by SETD7 seems to inhibit SMYD2-dependent p53K370 methylation through blocking the interaction between SMYD2 and p53 (16). But whether SMYD3 can modulate p53 expression remains unknown.

In the present study, we sought to determine whether and, if so, how SMYD3 regulates the growth of glioma. We found that SMYD3 was overexpressed in human glioma but not in normal brain tissue. Enforced SMYD3 expression promoted glioma cell proliferation. Inhibition of SMYD3 expression in glioma cells suppressed their anchorage-independent growth in vitro and tumorigenicity in vivo. Furthermore, we found that SMYD3 regulates the expression of p53 protein, which is essential in SMYD3-induced cell growth in glioma cells. These results showed that SMYD3 is overexpressed in human glioblastomas and contributes to glioma tumorigenicity.

Materials and methods

Samples, cells and antibodies

Human normal brain tissues and glioma tissue samples were obtained from patients who underwent therapeutic surgical procedures at the Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University. The adult normal brain tissues as normal controls were obtained from surgical resections of 18 trauma patients. Clinicopathological features and treatment strategies of all the glioma patients are shown in Table I. All experiments were approved by the Ethics Committee of Beijing Tian Tan Hospital and informed consent was obtained from all patients prior to specimen collection. Human glioma cell lines, HEB, U87, U373, LN-18 and T98G, were obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA). The cells were maintained in Dulbecco's modified Eagle's medium (DMEM; Gibco-BRL, Grand Island, NY, USA) supplemented with 10% fetal bovine serum (FBS) (Invitrogen) and cultured at 37°C, in 5% CO2 in a humidified chamber. Mouse monoclonal SMYD3, p53, p21 and β-actin antibodies were purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, uSA).

Table I

SMYD3 staining and clinicopathological characteristics of the 46 glioma patients.

Table I

SMYD3 staining and clinicopathological characteristics of the 46 glioma patients.

VariablesSMYD3 staining
TotalP-valuea
LowHigh
Age (years)
 ≤6079160.272
 >60161430
Gender
 Male1113240.579
 Female101222
Tumor diameter (cm)
 ≤3108180.463
 >3121628
Tumor locus
 Supratentorial1715320.311
 Infratentorial5914
TNM stage
 Low grades I and II8190.001b
 High grades III and IV73037

a χ2 test.

b Comparing TNM stages I–II vs. III–IV. SMYD3, SET and MYND domain-containing protein 3.

Plasmid construction and transfection

For overexpression, the cDNA representing the complete open reading frame of SMYD3 was cloned into the pBabe vector to generate the SMYD3 expression plasmid. The expression plasmid was verified by sequencing both strands and was used to transfect the LN-18 cells to establish the SMYD3 overexpression cell line. For SMYD3 RNA interference, the control (pSuper) and pSuper-sh SMYD3 plasmids were purchased from OligoEngine (Seattle, WA, USA) and were used to transfect the T98G cells to establish the SMYD3 knockdown cell line. The transfection efficiency of SMYD3 was confirmed by western blotting and quantitative reverse transcription-PCR (qRT-PCR) analyses.

MTT assay

A 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) assay was used to assess cell proliferation. The cells were seeded and 20 ml of the MTT solution (5 mg/ml) was then added to each well at the indicated time. The absorbance at 570 nm was measured using a microplate reader (Bio-Rad, Hercules, CA, USA).

Immunohistochemistry

Paraffin-embedded sections were deparaffinized, blocked and incubated with antibody at 4°C overnight. Horseradish peroxidase-conjugated secondary antibody (1:500) was then added and further incubated for 1 h at room temperature. The sections were developed using a 3,3′-diaminobenzidine tetrahydrochloride (DAB) substrate kit at room temperature for 1–5 min and then counterstained with hematoxylin. The extent of immunostaining was quantified by counting the percentage of positive cells and classified into four groups: 0, <25% positive cells; 1, 25–50% positive cells; 2, 51–75% positive cells; and 3, >75% positive cells (17). The immunohistochemistry score cut-off point was established as 30 using X-tile software program (version 3.6.3; Yale University School of Medicine, New Haven, CT, USA).

Colony formation assay

The cells were seeded in 6-cm dishes at a density of 300 cells/dish. After incubation for 14 days, the colonies were fixed with methanol for 10 min and stained with crystal violet for 15 min, after which point the number of colonies containing >50 cells was scored (18).

Western blot assay

Equal amounts of protein were separated using SDS polyacrylamide gels and were electrotransferred to polyvinylidene fluoride membranes (Millipore, Bedford, MA, USA). The membranes were immunoblotted overnight at 4°C with primary antibodies, followed by their respective secondary antibodies. β-actin was used as the loading control.

qRT-PCR

RNA was extracted using TRIzol reagent, according to the manufacturer's recommended instructions (Invitrogen). qRT-PCR was performed using Applied Biosystems (Foster City, CA, USA) StepOne and StepOne Plus real-Time PCR Systems. GAPDH was used as a loading control. The experiments were repeated a minimum of three times to confirm the results.

Immunofluorescence staining

The cells were grown on sterile coverslips, and then the cells were fixed with 4% paraformaldehyde and permeabilized using 0.1% Triton X-100. Cells were blocked with the primary antibody followed by rhodamine-conjugated anti-rabbit secondary antibody. Finally, the cells were further stained with 4′,6-diamidino-2-phenylindole (DAPI).

In vivo tumorigenesis assays

The in vivo tumorigenesis and metastasis assays were performed as previously described (19). Briefly, 1×106 cells were injected subcutaneously into the right flanks of severe combined immunodeficiency (SCID) mice. Tumor length (L) and width (W) were measured every 3 days, and tumor volume was calculated using the equation: Volume = (W2 × L)/2. After 5 weeks, the mice were sacrificed and the tumor volume and weight were measured. All of the animal experiments were performed with the approval of the Weifang People's Hospital Animal Care and Use Committee.

Statistical analysis

Experimental data are shown as mean ± standard deviation (SD). The results from different treatment groups were compared using a two-tailed Student's t-test. The Kaplan-Meier method was used to estimate the probability of patient survival, and differences in the survival of subgroups of patients were compared using Mantel's log-rank test. Differences were considered to be statistically significant at a value of P<0.05. Statistical analysis was carried out with SPSS/Win11.0 software (SPSS, Inc., Chicago, IL, USA).

Results

Expression of SMYD3 is directly correlated with the grade of glioma and is associated with decreased patient survival

We analyzed SMYD3 expression in human normal brain and glioma tissues by immunohistochemistry. No staining was detected in the normal human brain samples, and the SMYD3 protein was nearly expressed in all human glioma samples. The protein was localized in the nuclei of the tumor cells (Fig. 1A). The ratio of SMYD3-positive cells in the glioma samples was higher than that in the human normal brain samples (Fig. 1B). A high level of SMYD3 expression was significantly more common in glioma tissues with high pathologic grade than the level in tissues with low pathologic grade (P=0.001, Table I). No significant association was found between SMYD3 expression and gender, diameter, tumor locus or age at diagnosis. Kaplan-Meier survival analysis showed significant prolongation of survival in patients with SMYD3 low-expression gliomas compared with the survival of patients with SMYD3 high-expression gliomas (Fig. 2). The cut-off point was established using the X-tile software program. These results indicated that increased expression of SMYD3 was significantly associated with poor overall survival of the glioma patients.

Transfection efficiency of SMYD3 in glioma cell lines

We next analyzed the expression of SMYD3 protein in the immortalized normal human astrocyte cell line HEB and glioma cell lines U87, U373, LN-18 and T98G. Significantly higher expression of SMYD3 protein (Fig. 3A and B) and mRNA (Fig. 3C) was evident in the U87, U373, LN-18 and T98G glioma cell lines when compared with that in the immortalized normal human astrocyte HEB cells. The SMYD3 expression plasmid pBabe-SMYD3 was transfected into LN-18 cells. After selection with puromycin, SMYD3 expression was assayed by western blot analysis (Fig. 3D) and qRT-PCr (Fig. 3E). T98G is a highly tumorigenic human glioma cell line commonly used in glioma research. This cell line was infected with pSuper-shSMYD3 or the control (pSuper) to investigate the effect of SMYD3 knockdown on the proliferation and colony formation of glioma cells. Western blot analysis (Fig. 3F) and qRT-PCR (Fig. 3G) demonstrated that the protein and mRNA expression levels of SMYD3 were significantly suppressed in the cells infected with pSuper-shSMYD3.

SMYD3 promotes glioma cell proliferation in vitro

The effect of SMYD3 overexpression on cell proliferation was determined to characterize the functions of SMYD3 in glioma. The ability of the SMYD3-expressing LN-18 cells to form spheres was analyzed to determine the function of SMYD3 in cellular transformation. The colony formation assay revealed that SMYD3 overexpression significantly increased the number of colonies in the LN-18-pBabe-SMYD3 cells (Fig. 4A). The MTT assay showed that the LN-18-pBabe-SMYD3 cells displayed a higher proliferation rate than that of the control cells (Fig. 4B). The colony formation assay revealed that SMYD3 suppression significantly decreased the number of colonies (Fig. 4C). The results of the MTT assay also showed that SMYD3 suppression in the T98G cells was associated with decreased cell proliferation (Fig. 4D). As PCNA is an important marker of cell proliferation, we next examined PCNA by immunofluorescence staining. As shown in Fig. 5A, we found that the overexpression of SMYD3 in LN-18 cells significantly upregulated PCNA staining. In addition, knockdown of SMYD3 in T98G cells markedly downregulated the staining of PCNA (Fig. 5B). These results further confirmed that SMYD3 was involved in the proliferation of glioma cells.

Knockdown of SMYD3 inhibits glioma cell tumorigenesis in vivo

To explore the effects of SMYD3 on tumorigenesis in vivo, T98G-shSMYD3 and its control cells were injected subcutaneously into the flanks of nude mice. The diameters of the tumors were measured every 7 days. We found that the control cells formed tumors earlier and that the tumor volumes were much larger in those that were formed from the control cells than in those that were formed from the SMYD3 knockdown cells (Fig. 6A and B). As shown in Fig. 6C, the tumor weight of the T98G control was much higher than that of the SMYD3-knockdown cells. These results revealed that SMYD3 promotes glioma cell xenograft formation and growth in vivo.

Inactivation of p53 contributes to SMYD3-induced oncogenic activities

Previous studies have shown that p53 is important in glioma cell proliferation (20). Thus, this study investigated whether p53 is modulated by SMYD3 in glioma cells. We found that SMYD3 overexpression in LN-18 cells significantly decreased expression of p53 and its target protein p21 (Fig. 7A). In contrast, SMYD3 knockdown in T98G cells significantly increased the expression levels of p53 and p21 protein (Fig. 7A). These results suggest that SMYD3 promotes glioma cell growth partly through p53 inhibition.

To test whether p53 is involved in the SMYD3 induced proliferation, we determined the role of p53 in shSMYD3-induced proliferation inhibition by knocking down p53 expression using siRNA in T98G cells. The p53 knockdown efficiency was detected using western blot analysis at 48 h after transfection (Fig. 7B). As shown in Fig. 7C and D, the proliferation inhibition that was induced by shSMYD3 was obviously reversed following p53 knockdown using siRNA. To recognize any clinical correlation of SMYD3 and p53, we analyzed p53 expression in the same human glioma tissues. A highly negative correlation between SMYD3 and p53 expression was confirmed (Fig. 7E). This result was consistent with and further supports our above analysis. These results confirmed that p53 is involved in the SMYD3-mediated proliferation ability in glioma cells.

Discussion

Gliomas account for >50% of all primary brain tumors, and nearly two-thirds of gliomas are highly aggressive with 'malignant' pathological features (WHO grade III or IV) (2). Glioma cells are a mixture of heterogeneous cell populations, and numerous factors are likely to be involved in dictating recurrence, progression and patient survival (4). Despite advances in the neurosurgical, radiotherapeutic, and chemotherapeutic treatment of gliomas, the prognosis generally remains poor. This study aimed to characterize the functions of SMYD3 in glioma.

Previous results have shown that SMYD3 is expressed differentially in normal and cancer tissues (2124). SMYD3 overexpression has been associated with progression in gastric and breast cancer (23,25). However, the effect of altered SMYD3 expression on the progression of glioma cells remains elusive. This study examined the expression of SMYD3 in glioma and investigated the relationship between SMYD3 expression and the clinicopathological characteristics of patients with glioma. This study demonstrated that there was a significant difference in SMYD3 expression between glioma and the normal tissues. These results were similar to those of previous studies of other human cancers (26). Furthermore, high expression of SMYD3 in the gliomas was correlated with clinicopathological features, including tumor recurrence rate, histopathological classification and clinical stage. Overexpression of SMYD3 protein was thus correlated with poor prognosis of glioma patients. Overall survival rate was significantly different between the two groups, and these results showed that the higher the expression of SMYD3 protein, the lower the disease-free survival rate.

We also demonstrated that SMYD3 overexpression increased glioma cell proliferation and promoted tumorigenesis in vitro, whereas SMYD3 knockdown by shRNA inhibited tumorigenesis. These data are consistent with a recent study that investigated the function of SMYD3 in gastric and breast cancers (26,27). All of these findings are supported by a recent study, which reported that the overexpression of SMYD3 protein in breast cancer alters tumor cell fate and promotes tumor progression (27).

The initiation and progression of glioma involves a series of genetic events including activation of oncogenes and inactivation of tumor suppressors (28,29). Genetic studies have also shown that p53 is a central regulator of cell growth, proliferation and apoptosis (20). In this study, SMYD3 over-expression significantly decreased p53 expression, whereas SMYD3 knockdown produced an opposite effect in glioma cells. The inhibition of p53 by siRNA reversed the induction of shSMYD3 on glioma cell proliferation inhibition. Previous studies showed that SMYD2 monomethylates lysine 370 of p53; this lysine residue is located in the regulatory domain at the extreme carboxyl terminus of p53 (16). Knockdown of SMYD2 by small interfering RNA was found to enhance p53-mediated apoptosis in cancer cells (16). In addition, SMYD2-dependent p53K370 methylation impaired the expression of CDKN1A, an important downstream target of p53, implying that SMYD2 represses the function of p53 through K370 monomethylation. Thus, we hypothesized that the SMYD3 action may be through monomethylation of p53. However, the mechanisms involved in the expression profile of SMYD3 require further investigation. Additional studies are required to explore the relationships between the SMYD3 gene and other genes, and its relationship to other molecules that may be associated with glioma.

In conclusion, this study demonstrated for the first time that SMYD3 is overexpressed in glioma tissues. SMYD3 over-expression increased the growth of glioma cells in vitro and promoted glioma tumorigenesis in vivo. Therefore, SMYD3 is potentially an important molecular target for novel anti-glioma therapy.

Acknowledgments

This study was supported by the National Key Technology Research and Development Program of the Ministry of Science and Technology of China (no. 2013BAI09B03).

References

1 

Cloughesy TF, Cavenee WK and Mischel PS: Glioblastoma: From molecular pathology to targeted treatment. Annu Rev Pathol. 9:1–25. 2014. View Article : Google Scholar

2 

DeSantis CE, Lin CC, Mariotto AB, Siegel RL, Stein KD, Kramer JL, Alteri R, Robbins AS and Jemal A: Cancer treatment and survivorship statistics, 2014. CA Cancer J Clin. 64:252–271. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Annibali D, Whitfield JR, Favuzzi E, Jauset T, Serrano E, Cuartas I, Redondo-Campos S, Folch G, Gonzàlez-Juncà A, Sodir NM, et al: Myc inhibition is effective against glioma and reveals a role for Myc in proficient mitosis. Nat Commun. 5:46322014. View Article : Google Scholar : PubMed/NCBI

4 

Ahmed R, Oborski MJ, Hwang M, Lieberman FS and Mountz JM: Malignant gliomas: Current perspectives in diagnosis, treatment, and early response assessment using advanced quantitative imaging methods. Cancer Manag Res. 6:149–170. 2014.PubMed/NCBI

5 

Martin C and Zhang Y: Mechanisms of epigenetic inheritance. Curr Opin Cell Biol. 19:266–272. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Zhou VW, Goren A and Bernstein BE: Charting histone modifications and the functional organization of mammalian genomes. Nat Rev Genet. 12:7–18. 2011. View Article : Google Scholar

7 

Van Aller GS, Reynoird N, Barbash O, Huddleston M, Liu S, Zmoos AF, McDevitt P, Sinnamon R, Le B, Mas G, et al: Smyd3 regulates cancer cell phenotypes and catalyzes histone H4 lysine 5 methylation. Epigenetics. 7:340–343. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Cock-Rada AM, Medjkane S, Janski N, Yousfi N, Perichon M, Chaussepied M, Chluba J, Langsley G and Weitzman JB: SMYD3 promotes cancer invasion by epigenetic upregulation of the metalloproteinase MMP-9. Cancer Res. 72:810–820. 2012. View Article : Google Scholar :

9 

He C, Xu J, Zhang J, Xie D, Ye H, Xiao Z, Cai M, Xu K, Zeng Y, Li H, et al: High expression of trimethylated histone H3 lysine 4 is associated with poor prognosis in hepatocellular carcinoma. Hum Pathol. 43:1425–1435. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Liu L, Kimball S, Liu H, Holowatyj A and Yang ZQ: Genetic alterations of histone lysine methyltransferases and their significance in breast cancer. Oncotarget. 6:2466–2482. 2015. View Article : Google Scholar :

11 

Liu Y, Deng J, Luo X, Pan Y, Zhang L, Zhang R and Liang H: Overexpression of SMYD3 was associated with increased STAT3 activation in gastric cancer. Med Oncol. 32:4042015. View Article : Google Scholar

12 

Luo XG, Xi T, Guo S, Liu ZP, Wang N, Jiang Y and Zhang TC: Effects of SMYD3 overexpression on transformation, serum dependence, and apoptosis sensitivity in NIH3T3 cells. IUBMB Life. 61:679–684. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Xu LX, Li ZH, Tao YF, Li RH, Fang F, Zhao H, Li G, Li YH, Wang J, Feng X, et al: Histone acetyltransferase inhibitor II induces apoptosis in glioma cell lines via the p53 signaling pathway. J Exp Clin Cancer Res. 33:1082014. View Article : Google Scholar : PubMed/NCBI

14 

Cohen AL and Colman H: Glioma biology and molecular markers. Cancer Treat Res. 163:15–30. 2015. View Article : Google Scholar

15 

Chuikov S, Kurash JK, Wilson JR, Xiao B, Justin N, Ivanov GS, McKinney K, Tempst P, Prives C, Gamblin SJ, et al: Regulation of p53 activity through lysine methylation. Nature. 432:353–360. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Huang J, Perez-Burgos L, Placek BJ, Sengupta R, Richter M, Dorsey JA, Kubicek S, Opravil S, Jenuwein T and Berger SL: repression of p53 activity by Smyd2-mediated methylation. Nature. 444:629–632. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Wang q, Wang Y, Zhang Y, Zhang Y and Xiao W: The role of uPAR in epithelial-mesenchymal transition in small airway epithelium of patients with chronic obstructive pulmonary disease. Respir Res. 14:672013. View Article : Google Scholar : PubMed/NCBI

18 

Sun Y, Wang Y, Fan C, Gao P, Wang X, Wei G and Wei J: Estrogen promotes stemness and invasiveness of ER-positive breast cancer cells through Gli1 activation. Mol Cancer. 13:1372014. View Article : Google Scholar : PubMed/NCBI

19 

Wang Y, Wen M, Kwon Y, Xu Y, Liu Y, Zhang P, He X, Wang Q, Huang Y, Jen KY, et al: CUL4A induces epithelial-mesenchymal transition and promotes cancer metastasis by regulating ZEB1 expression. Cancer Res. 74:520–531. 2014. View Article : Google Scholar :

20 

Zheng H, Ying H, Yan H, Kimmelman AC, Hiller DJ, Chen AJ, Perry SR, Tonon G, Chu GC, Ding Z, et al: p53 and Pten control neural and glioma stem/progenitor cell renewal and differentiation. Nature. 455:1129–1133. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Wang SZ, Luo XG, Shen J, Zou JN, Lu YH and Xi T: Knockdown of SMYD3 by RNA interference inhibits cervical carcinoma cell growth and invasion in vitro. BMB Rep. 41:294–299. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Sponziello M, Durante C, Boichard A, Dima M, Puppin C, Verrienti A, Tamburrano G, Di Rocco G, Redler A, Lacroix L, et al: Epigenetic-related gene expression profile in medullary thyroid cancer revealed the overexpression of the histone methyltransferases EZH2 and SMYD3 in aggressive tumours. Mol Cell Endocrinol. 392:8–13. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Liu Y, Liu H, Luo X, Deng J, Pan Y and Liang H: Overexpression of SMYD3 and matrix metalloproteinase-9 are associated with poor prognosis of patients with gastric cancer. Tumour Biol. Jan 28–2015.Epub ahead of print. View Article : Google Scholar

24 

Suzuki S, Nozawa Y, Tsukamoto S, Kaneko T, Imai H and Minami N: Histone methyltransferase Smyd3 regulates early embryonic lineage commitment in mice. Reproduction. 150:21–30. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Hamamoto R, Silva FP, Tsuge M, Nishidate T, Katagiri T, Nakamura Y and Furukawa Y: Enhanced SMYD3 expression is essential for the growth of breast cancer cells. Cancer Sci. 97:113–118. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Ren TN, Wang JS, He YM, Xu CL, Wang SZ and Xi T: Effects of SMYD3 over-expression on cell cycle acceleration and cell proliferation in MDA-MB-231 human breast cancer cells. Med Oncol. 28(Suppl 1): S91–S98. 2011. View Article : Google Scholar

27 

Liu Y, Luo X, Deng J, Pan Y, Zhang L and Liang H: SMYD3 overexpression was a risk factor in the biological behavior and prognosis of gastric carcinoma. Tumour Biol. 36:2685–2694. 2015. View Article : Google Scholar

28 

Xu Y, Wang Y, Ma G, Wang Q and Wei G: CUL4A is overexpressed in human pituitary adenomas and regulates pituitary tumor cell proliferation. J Neurooncol. 116:625–632. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Wang Y, Ma G, Wang Q, Wen M, Xu Y, He X, Zhang P, Wang Y, Yang T, Zhan P, et al: Involvement of CUL4A in regulation of multidrug resistance to P-gp substrate drugs in breast cancer cells. Molecules. 19:159–176. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2015
Volume 34 Issue 5

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
Dai B, Wan W, Zhang P, Zhang Y, Pan C, Meng G, Xiao X, Wu Z, Jia W, Zhang J, Zhang J, et al: SET and MYND domain-containing protein 3 is overexpressed in human glioma and contributes to tumorigenicity. Oncol Rep 34: 2722-2730, 2015
APA
Dai, B., Wan, W., Zhang, P., Zhang, Y., Pan, C., Meng, G. ... Zhang, L. (2015). SET and MYND domain-containing protein 3 is overexpressed in human glioma and contributes to tumorigenicity. Oncology Reports, 34, 2722-2730. https://doi.org/10.3892/or.2015.4239
MLA
Dai, B., Wan, W., Zhang, P., Zhang, Y., Pan, C., Meng, G., Xiao, X., Wu, Z., Jia, W., Zhang, J., Zhang, L."SET and MYND domain-containing protein 3 is overexpressed in human glioma and contributes to tumorigenicity". Oncology Reports 34.5 (2015): 2722-2730.
Chicago
Dai, B., Wan, W., Zhang, P., Zhang, Y., Pan, C., Meng, G., Xiao, X., Wu, Z., Jia, W., Zhang, J., Zhang, L."SET and MYND domain-containing protein 3 is overexpressed in human glioma and contributes to tumorigenicity". Oncology Reports 34, no. 5 (2015): 2722-2730. https://doi.org/10.3892/or.2015.4239