TET1 partially mediates HDAC inhibitor-induced suppression of breast cancer invasion

  • Authors:
    • Hou-Gen Lu
    • Wang Zhan
    • Lin Yan
    • Rui-Ying Qin
    • Yi-Peng Yan
    • Zhen-Jiang Yang
    • Gui‑Chao  Liu
    • Gui-Qin Li
    • Hai-Feng Wang
    • Xing-Liang Li
    • Zhi Li
    • Lu Gao
    • Guo-Qing Chen
  • View Affiliations

  • Published online on: August 26, 2014     https://doi.org/10.3892/mmr.2014.2517
  • Pages: 2595-2600
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Histone deacetylases (HDACs) are important in chromatin remodeling and epigenetic regulation of gene expression. Histone deacetylase inhibitors (HDACi) have highly effective anti-metastatic and anti-angiogenic activity in various types of cancer, while the molecular mechanisms involved in this process are not fully understood. In the present study, trichostatin A (TSA), a HDACi, was found to suppress MCF-7 breast carcinoma cell invasion and upregulate TET1 expression in a dose-dependent manner. TET1, a dioxygenase involved in cytosine demethylation, is downregulated during breast cancer progression. TET1 knockdown in MCF-7 cells facilitates cell invasion, inhibits the expression of tissue inhibitors of metalloproteinase 2/3 (TIMP2/3) and promotes matrix metalloproteinases (MMP) 2/9 transcriptional activity. Importantly, TET1 depletion impaired the inhibitory effect of TSA on breast cancer cell invasion. Together, these results illustrated a mechanism by which TET1 partially mediates HDACi elicited suppression of breast cancer invasion.

Introduction

Tumor invasion and metastasis are the main characteristics of various types of aggressive human cancer, including breast cancer (1). Cell invasion is one of the initiation steps for the metastatic cascade, during which cancer cells migrate through the extracellular matrix from the primary tumor, which is associated with the upregulated expression of matrix metalloproteinases (MMPs) (24). Therefore, it is important to elucidate the mechanisms underlying cancer invasion.

Histone acetylation status regulated by histone acetyltransferases (HATs) and histone deacetylases (HDACs) is important in the regulation of gene expression by affecting chromatin structure and accessibility (5,6). HDACs are recruited to DNA-bound transcription factors resulting in the removal of acetyl groups from nucleosomal histones or directly interact with transcription factors to modulate gene expression (710). HDAC inhibition leads to the accumulation of acetylation in histones and transcription factors, and specifically programmed gene expression patterns (11,12).

In humans, the reduction of histone acetylation is significantly associated with tumor progression and invasion (13). Emerging evidence indicates that histone deacetylase inhibitors (HDACi) induce growth inhibition, cell cycle arrest and programmed cell death in diverse cancer cells (14,15). HDACi treatment upregulates the expression of suppressors of metastasis and downregulates invasion-promoting genes, resulting in the repression of cancer cell invasion and metastasis (16). HDAC inhibition is emerging as a potential strategy for cancer therapy and several HDACi have been developed for clinical trials in patients with solid malignancies (17). Trichostatin A (TSA), a non-competitive reversible inhibitor of HDAC activity, has been reported to inhibit cancer invasion and metastasis in vivo and in vitro (1820). HDACi modulate cancer progression through affecting the acetylation of histone and non-histone proteins to reactivate the transcription of differential target genes. It has been proposed that TSA upregulates RECK to suppress MMP2 activation and cancer cell invasion (21). Therefore, it is important to investigate the targets of HDACi in order to elucidate the molecular mechanisms underlying HDACi elicited phenotypes.

In the present study, TSA was observed to inhibit cell invasion in MCF-7 breast cancer cells. Furthermore, the expression of tissue inhibitors of metalloproteinase 2/3 (TIMP2/3) was upregulated and the expression of MMP2/9 was decreased by TSA treatment. Notably, TIMP2/3 and MMP2/9 have been revealed as targets of TET1 in prostate and breast cancer invasion (22). Additionally, tumor development is associated with a decrease in TET expression and 5-methylcytosine hydroxylation (23). TIMP2 and TET1 consistently demonstrated downregulation during breast cancer progression in vivo. Our hypothesis was that TET1 may be one of the targets of HDACi in breast cancer invasion. As expected, TET1 was upregulated by TSA stimulation and TET1 knockdown facilitated breast cancer cell invasion. Importantly, TET1 depletion impaired TSA induced suppression of cell invasion, suggesting that TET1 may act as one of the HDACi targets partially mediating TSA elicited anti-cancer activity.

Materials and methods

Patient samples and cell culture

The present study was approved by the ethics committee of Shanghai Tongren Hospital (Shanghai, China), and written informed consent was obtained from all participants prior to the study. A total of 61 cancer specimens from breast cancer patients from stages I to IV were collected and the conditions of these patients are summarized in Table I. Samples were individually fresh-frozen in TRIzol reagent (Invitrogen Life Technologies, Carlsbad, CA, USA). All patients were histologically examined at Shanghai Tongren Hospital (Shanghai, China) and written informed consent was obtained from all study participants. Breast cancer MCF-7 cells were cultured in RPMI-1640 medium (Invitrogen Life Technologies) supplemented with 10% fetal calf serum at 37°C and 5% CO2. TSA dissolved in DMSO was applied in the present study with the indicated concentrations.

Table I

Characteristics of breast cancer patients.

Table I

Characteristics of breast cancer patients.

Age (years)

Phase40–4950+
I57
II1214
III79
IV34
Wound-healing assay

Wound healing was performed as previously described in 12-well plates (24). Briefly, after MCF-7 cells grew to >90% confluence, a wound was introduced using a 200 μl pipette tip. Subsequently, the cells were washed once with PBS and then incubated at 37°C and 5% CO2. Wound closure was monitored over an indicated time period and images were captured at the four intersecting edges of the cross. Wound width at 0 and 24 or 36 h was measured and the difference plotted as the percentage of wound closure.

Quantitative real-time PCR (qRT-PCR)

Total RNA was extracted from MCF-7 cells or cancer tissues using TRIzol reagent (Invitrogen Life Technologies). For each sample, 2.5 or 5 μg of total RNA was reverse transcribed using the QuantiTect Reverse Transcription kit (Qiagen, Hilden, Germany). The cDNA product was then quantified by SYBR-Green real-time PCR master mix (Toyobo, Osaka, Japan). The primers used were as follows: TET1, forward 5′-GAGCCTGTTCCTCGATGTGG-3′ and reverse 5′-CAAACCCACCTGAGGCTGTT-3′; GAPDH, forward 5′-GTGTTCCTACCCCCAATGTGT-3′ and reverse 5′-ATTGTCATACCAGGAAATGAGCTT-3′; TIMP2, forward 5′-GGGTCTCGCTGGACATTG-3′ and reverse 5′-TTGATGTTCTTCTCCGTGACC-3′; TIMP3, forward 5′-CATGTGCAGTACATCCATACGG-3′ and reverse 5′-CATCATAGACGCGACCTGTCA-3′; MMP2, forward 5′-AAGGCCAAGTGGTCCGTGTGAA-3′ and reverse 5′-AACAGTGGACATGGCGGTCTCAG-3′; MMP9, forward 5′-CACGTCCACCCCTCAGAGC-3′ and reverse 5′-GCCACTTGTCGGCGATAAGC-3′.

Western blot analysis

MCF-7 cells were lysed with two-fold loading buffer [20 mM of Tris-HCl (pH 7.4), 2 mM of EDTA and 1% Triton X-100] and the supernatant was subjected to western blot analysis, which was performed as previously described (25). GAPDH was used as the loading control. The following primary antibodies were used: anti-TET1 (Abcam, Cambridge, UK; 1:500), anti-MMP2 (Abcam; 1:400), anti-TIMP2 (Abcam; 1:500) and anti-GAPDH (Millipore, Billerica, MA, USA; 1:10,000).

shRNA and transfection

Chemically synthesized TET1 shRNA and scramble shRNA (control) were annealed and cloned into a short interfering RNA expressing vector named pSUPER (Oligoengine, Seattle, WA, USA). Control or TET1 shRNA targeted sequences were: control shRNA 5′-GCTACGAAGCACCTCTCTTAG-3′ and TET1 shRNA 5′-CGATGCAAGCCATCCTTTCGA-3′. Plasmids purified by the Qiagen purification kit were transfected into MCF-7 cells at 40–60% confluency with Lipofectamine 2000 (Invitrogen Life Technologies) according to the manufacturer’s instructions.

Statistic analysis

SPSS 11.0 software was used for statistical analysis in the present study. The values are presented as the mean ± SD and one-way ANOVA was applied for group differences. All tests were two-sided and P<0.05 was considered to indicate a statistically significant difference.

Results

HDACi suppresses breast cancer cell invasion and regulates TIMP2/3 and MMP2/9 expression

Previously, HDACi were reported to inhibit breast cancer cell invasion (18,26). Prior to examining the mechanisms of HDAC in cancer invasion, the effects of TSA on cell invasion were examined by a wound-healing assay in MCF-7 breast cancer cells. Consistent with previous findings, TSA significantly suppressed cell invasion (Fig. 1A and B). TIMP2 and 3, the important suppressors of cancer cell invasion (27,28), were upregulated under the stimulation of TSA. By contrast, the expression of MMP2/9, which was correlated with clinicopathological disease variables in breast cancer (29,30), was increased by HDAC inhibition (Fig. 1C). Furthermore, TSA affected MMP2 and TIMP2 expression and this was verified at the protein level (Fig. 1D). In addition, MMP2/9 and TIMP2/3 mRNA levels were not affected when TSA was administered to cells within 2 h (data not shown). This demonstrates that TIMP2/3 and MMP2/9 expression may be indirectly modulated by HDAC in breast cancer.

TIMP2, MMP2 and TET1 expression during breast cancer development

Coincidently, TIMPs and MMPs were revealed as downstream targets of TET1 and TET1 suppresses invasion partly through TIMP activation and MMP inhibition in prostate and breast cancer (22). In order to investigate the correlation between TIMP2, MMP2 and TET1 in vivo, the expression of these genes in multiple breast cancer patients at different stages was examined by qRT-PCR. TIMP2 and MMP2 expression was progressively downregulated and upregulated with breast cancer development, respectively (Fig. 2A and B). Consistently, TET1 mRNA level was reduced in breast cancer tissues (Fig. 2C), which corrrelated with TIMP2 expression changes (23). Collectively, TIMP2 and TET1 downregulation and MMP2 upregulation were correlated with breast cancer progression.

TET1 knockdown facilitates breast cancer cell invasion

Subsequently, the present study examined whether the reduction of TET1 is functionally involved in breast cancer cell invasion. TET1 shRNA was delivered into MCF-7 cells to efficiently knockdown TET1 expression (Fig. 3A). Then the effects of TET1 knockdown on cell invasion were analyzed and the results demonstrated that TET1 depletion by TET1 shRNA increased the cell invasion capacity (Fig. 3B and C). Notably, TET1 knockdown resulted in a decrease in TIMP2/3 and the upregulation of MMP2/9 expression (Fig. 3D), which is opposite to the effects of TSA in MCF-7 cells (Fig. 1A). This indicates that TET1 suppresses breast cancer cell invasion.

TSA upregulates TET1 expression in MCF-7 cells

Given that the expression of TIMPs and MMPs was affected by HDAC inhibition and TET1, our hypothesis was that TET1 may be regulated by HDACi. As expected, TET1 expression was upregulated by TSA in a dose-dependent manner (Fig. 4A), which was confirmed by western blot analysis (Fig. 4B). Additionally, TET1 was responsive to short-term TSA treatment (Fig. 4C). This suggests that TET1 may be a target of TSA in breast cancer.

TET1 knockdown impairs TSA-induced suppression of breast cancer cell invasion

Since TET1 expression was found to be regulated by HDAC inhibition, the functional association between TSA and TET1 needed to be elucidated. In order to examine whether TET1 is able to mediate TSA induced suppression of breast cancer cells, TSA was subjected to TET1-knockdown and cell invasion was examined. It was revealed that the inhibitory effect of TSA on breast cancer cell invasion was impaired in TET1-knockdown cells when compared with control shRNA expressing cells (Fig. 5A and B). Correspondingly, TSA upregulated TIMP2/3 and downregulated MMP2/9 expression, which were also impaired in TET1-knockdown cells. This indicates that TET1 partially mediates TSA induced repression of breast cancer cell invasion.

Discussion

Breast cancer is one of the most common types of cancer and has a high risk of mortality among females all over the world (31). Therefore there is a great need for understanding the molecular mechanisms underlying cancer progression and for the development of more effective therapeutic strategies for breast cancer. Carcinogenesis is able to be regulated by genetic and epigenetic alterations and epigenetic alterations are a reversible process, which makes epigenetic modifications preferable for clinical therapy (32,33).

DNA methylation and histone modifications are the most important epigenetic mediators of transcriptional regulation and multiple HDACi are in clinical development for hematological and solid tumor treatment (34,35). HDACi result in the accumulation of acetylation in histones and non-histone proteins to modulate target gene expression in cancerous cells, which reactivates the expression of tumor suppressors (35). In the present study (Fig. 1) and previous studies (18,36), the non-competitive reversible HDACi TSA has been demonstrated to suppress breast cancer cell invasion. The limitations of HDAC inhibition in cancer therapy is the non-specificity and toxicity of the chemical inhibitors. It is important to understand the molecular mechanisms in HDACi treatment. The HDACi valproic acid (VPA) induces ERα expression in its anti-tumor effects (37) and TSA enhances the acetylation and stability of the ERα and p300 proteins that may contribute to breast cancer treatment (38). TSA also directly upregulates p53 and RECK, which are important in tumor suppression (21,39).

The present study found that TIMP2/3 were increased and that MMP2/9 were decreased when treated with TSA (Fig. 1). It has been proposed that TET1 suppresses breast cancer invasion through the activation of TIMPs and the inhibition of MMPs (22). Additionally, TET1 and TIMP2/3 reduction was inversely correlated with MMP2/9 upregulation during breast cancer progression (Fig. 2) (23). Notably, TET1 demonstrated similar functions in MCF-7 breast cancer cell invasion (Fig. 3) (22). It is reasonable to postulate that TSA may inhibit breast cancer invasion through the regulation of TET1 expression. Notably, TSA promoted TET1 expression in a dose-dependent manner (Fig. 4) and TET1 partially mediated TSA elicited suppression of cell invasion (Fig. 5), which was in accordance with our hypothesis. TET1 expression may be negatively controlled by a specific HDAC and TSA treatment releases the HDAC inhibition on TET1 expression. TET1 was found to be associated with TIMPs genes and promoter regions to regulate their DNA methylation status and transcription levels (22,40). Our hypothesis is that TSA may indirectly affect 5-methylcytosine hydroxylation of TIMP gene promoters through the regulation of TET1 expression, which will be investigated in our future study. There is a possibility that VPA enhances the global 5-methylcytosine level in nuclear DNA (41).

Furthermore, the alteration in DNA methylation status is another important epigenetic modification and DNA methyltransferases (DNMTs) have become an epigenetic therapy target in various types of cancer (42). 5-azacytidine, a global DNMT inhibitor, has been approved for clinical trials against solid tumors (43). Currently, synergistic treatment with DNMT and HDACi has been applied for producing optimal effects (41).

As summarized in Fig. 5D, the HDACi TSA may upregulate the expression of TET1, which results in the activation of TIMPs and the inhibition of MMPs, thus, leading to the suppression of breast cancer cell invasion. The present study provides a novel molecular mechanism for HDAC inhibition in tumor suppression. This may provide insights into the role of HDACs in cancer development and epigenetic therapy.

Acknowledgements

We would like to thank Shanghai Xuhai Biological Technology Co., Ltd for the experimental support.

References

1 

Steeg PS: Metastasis suppressors alter the signal transduction of cancer cells. Nat Rev Cancer. 3:55–63. 2003. View Article : Google Scholar : PubMed/NCBI

2 

Mohanam S, Sawaya R, McCutcheon I, Ali-Osman F, Boyd D and Rao JS: Modulation of in vitro invasion of human glioblastoma cells by urokinase-type plasminogen activator receptor antibody. Cancer Res. 53:4143–4147. 1993.PubMed/NCBI

3 

Rao JS: Molecular mechanisms of glioma invasiveness: the role of proteases. Nat Rev Cancer. 3:489–501. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Davies B, Waxman J, Wasan H, Abel P, Williams G, Krausz T, Neal D, Thomas D, Hanby A and Balkwill F: Levels of matrix metalloproteases in bladder cancer correlate with tumor grade and invasion. Cancer Res. 53:5365–5369. 1993.PubMed/NCBI

5 

Horikoshi M: Histone acetylation: from code to web and router via intrinsically disordered regions. Curr Pharm Des. 19:5019–5042. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Gräff J and Tsai LH: Histone acetylation: molecular mnemonics on the chromatin. Nat Rev Neurosci. 14:97–111. 2013.PubMed/NCBI

7 

Mayo MW, Denlinger CE, Broad RM, Yeung F, Reilly ET, Shi Y and Jones DR: Ineffectiveness of histone deacetylase inhibitors to induce apoptosis involves the transcriptional activation of NF-kappa B through the Akt pathway. J Biol Chem. 278:18980–18989. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Brochier C, Dennis G, Rivieccio MA, McLaughlin K, Coppola G, Ratan RR and Langley B: Specific acetylation of p53 by HDAC inhibition prevents DNA damage-induced apoptosis in neurons. J Neurosci. 33:8621–8632. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Sachweh MC, Drummond CJ, Higgins M, Campbell J and Laín S: Incompatible effects of p53 and HDAC inhibition on p21 expression and cell cycle progression. Cell Death Dis. 4:e5332013. View Article : Google Scholar : PubMed/NCBI

10 

Cheung WL, Briggs SD and Allis CD: Acetylation and chromosomal functions. Curr Opin Cell Biol. 12:326–333. 2000. View Article : Google Scholar : PubMed/NCBI

11 

Van Lint C, Emiliani S and Verdin E: The expression of a small fraction of cellular genes is changed in response to histone hyperacetylation. Gene Expr. 5:245–253. 1996.PubMed/NCBI

12 

Mitsiades CS, Mitsiades NS, McMullan CJ, et al: Transcriptional signature of histone deacetylase inhibition in multiple myeloma: biological and clinical implications. Proc Natl Acad Sci USA. 101:540–545. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Song J, Noh JH, Lee JH, et al: Increased expression of histone deacetylase 2 is found in human gastric cancer. APMIS. 113:264–268. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Shao Y, Gao Z, Marks PA and Jiang X: Apoptotic and autophagic cell death induced by histone deacetylase inhibitors. Proc Natl Acad Sci USA. 101:18030–18035. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Marks PA, Richon VM, Miller T and Kelly WK: Histone deacetylase inhibitors. Adv Cancer Res. 91:137–168. 2004. View Article : Google Scholar : PubMed/NCBI

16 

McGarry LC, Winnie JN and Ozanne BW: Invasion of v-Fos(FBR)-transformed cells is dependent upon histone deacetylase activity and suppression of histone deacetylase regulated genes. Oncogene. 23:5284–5292. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Marks PA, Richon VM, Kelly WK, Chiao JH and Miller T: Histone deacetylase inhibitors: development as cancer therapy. Novartis Found Symp. 259:269–281. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Vigushin DM, Ali S, Pace PE, et al: Trichostatin A is a histone deacetylase inhibitor with potent antitumor activity against breast cancer in vivo. Clin Cancer Res. 7:971–976. 2001.

19 

Tarasenko N, Nudelman A, Tarasenko I, et al: Histone deacetylase inhibitors: the anticancer, antimetastatic and antiangiogenic activities of AN-7 are superior to those of the clinically tested AN-9 (Pivanex). Clin Exp Metastasis. 25:703–716. 2008. View Article : Google Scholar

20 

Bolden JE, Peart MJ and Johnstone RW: Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov. 5:769–784. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Liu LT, Chang HC, Chiang LC and Hung WC: Histone deacetylase inhibitor up-regulates RECK to inhibit MMP-2 activation and cancer cell invasion. Cancer Res. 63:3069–3072. 2003.PubMed/NCBI

22 

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

23 

Yang H, Liu Y, Bai F, et al: Tumor development is associated with decrease of TET gene expression and 5-methylcytosine hydroxylation. Oncogene. 32:663–669. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Asrani K, Keri RA, Galisteo R, et al: The HER2- and heregulin beta1 (HRG)-inducible TNFR superfamily member Fn14 promotes HRG-driven breast cancer cell migration, invasion, and MMP9 expression. Mol Cancer Res. 11:393–404. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Brown SA, Ghosh A and Winkles JA: Full-length, membrane-anchored TWEAK can function as a juxtacrine signaling molecule and activate the NF-kappaB pathway. J Biol Chem. 285:17432–17441. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Grimaldi C, Pisanti S, Laezza C, et al: Anandamide inhibits adhesion and migration of breast cancer cells. Exp Cell Res. 312:363–373. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Albini A, Melchiori A, Santi L, Liotta LA, Brown PD and Stetler-Stevenson WG: Tumor cell invasion inhibited by TIMP-2. J Natl Cancer Inst. 83:775–779. 1991. View Article : Google Scholar : PubMed/NCBI

28 

Baker AH, George SJ, Zaltsman AB, Murphy G and Newby AC: Inhibition of invasion and induction of apoptotic cell death of cancer cell lines by overexpression of TIMP-3. Br J Cancer. 79:1347–1355. 1999. View Article : Google Scholar : PubMed/NCBI

29 

Jinga D, Stefanescu M, Blidaru A, Condrea I, Pistol G and Matache C: Serum levels of matrix metalloproteinases MMP-2 and MMP-9 and their tissue natural inhibitors in breast tumors. Roum Arch Microbiol Immunol. 63:141–158. 2004.PubMed/NCBI

30 

Sier CF, Kubben FJ, Ganesh S, et al: Tissue levels of matrix metalloproteinases MMP-2 and MMP-9 are related to the overall survival of patients with gastric carcinoma. Br J Cancer. 74:413–417. 1996. View Article : Google Scholar

31 

Jemal A, Siegel R, Ward E, et al: Cancer statistics, 2008. CA Cancer J Clin. 58:71–96. 2008. View Article : Google Scholar

32 

Taby R and Issa JP: Cancer epigenetics. CA Cancer J Clin. 60:376–392. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Cai FF, Kohler C, Zhang B, Wang MH, Chen WJ and Zhong XY: Epigenetic therapy for breast cancer. Int J Mol Sci. 12:4465–4487. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Namdar M, Perez G, Ngo L and Marks PA: Selective inhibition of histone deacetylase 6 (HDAC6) induces DNA damage and sensitizes transformed cells to anticancer agents. Proc Natl Acad Sci USA. 107:20003–20008. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Liu T, Kuljaca S, Tee A and Marshall GM: Histone deacetylase inhibitors: multifunctional anticancer agents. Cancer Treat Rev. 32:157–165. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Yang X, Phillips DL, Ferguson AT, Nelson WG, Herman JG and Davidson NE: Synergistic activation of functional estrogen receptor (ER)-alpha by DNA methyltransferase and histone deacetylase inhibition in human ER-alpha-negative breast cancer cells. Cancer Res. 61:7025–7029. 2001.

37 

Travaglini L, Vian L, Billi M, Grignani F and Nervi C: Epigenetic reprogramming of breast cancer cells by valproic acid occurs regardless of estrogen receptor status. Int J Biochem Cell Biol. 41:225–234. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Kim SH, Kang HJ, Na H and Lee MO: Trichostatin A enhances acetylation as well as protein stability of ERalpha through induction of p300 protein. Breast Cancer Res. 12:R222010. View Article : Google Scholar

39 

Henderson C, Mizzau M, Paroni G, Maestro R, Schneider C and Brancolini C: Role of caspases, Bid, and p53 in the apoptotic response triggered by histone deacetylase inhibitors trichostatin-A (TSA) and suberoylanilide hydroxamic acid (SAHA). J Biol Chem. 278:12579–12589. 2003. View Article : Google Scholar

40 

Williams K, Christensen J, Pedersen MT, et al: TET1 and hydroxymethylcytosine in transcription and DNA methylation fidelity. Nature. 473:343–348. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Kristensen LS, Nielsen HM and Hansen LL: Epigenetics and cancer treatment. Eur J Pharmacol. 625:131–142. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Hatziapostolou M and Iliopoulos D: Epigenetic aberrations during oncogenesis. Cell Mol Life Sci. 68:1681–1702. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Chik F and Szyf M: Effects of specific DNMT gene depletion on cancer cell transformation and breast cancer cell invasion; toward selective DNMT inhibitors. Carcinogenesis. 32:224–232. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2014
Volume 10 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
Lu H, Zhan W, Yan L, Qin R, Yan Y, Yang Z, Liu GC, Li G, Wang H, Li X, Li X, et al: TET1 partially mediates HDAC inhibitor-induced suppression of breast cancer invasion. Mol Med Rep 10: 2595-2600, 2014
APA
Lu, H., Zhan, W., Yan, L., Qin, R., Yan, Y., Yang, Z. ... Chen, G. (2014). TET1 partially mediates HDAC inhibitor-induced suppression of breast cancer invasion. Molecular Medicine Reports, 10, 2595-2600. https://doi.org/10.3892/mmr.2014.2517
MLA
Lu, H., Zhan, W., Yan, L., Qin, R., Yan, Y., Yang, Z., Liu, G., Li, G., Wang, H., Li, X., Li, Z., Gao, L., Chen, G."TET1 partially mediates HDAC inhibitor-induced suppression of breast cancer invasion". Molecular Medicine Reports 10.5 (2014): 2595-2600.
Chicago
Lu, H., Zhan, W., Yan, L., Qin, R., Yan, Y., Yang, Z., Liu, G., Li, G., Wang, H., Li, X., Li, Z., Gao, L., Chen, G."TET1 partially mediates HDAC inhibitor-induced suppression of breast cancer invasion". Molecular Medicine Reports 10, no. 5 (2014): 2595-2600. https://doi.org/10.3892/mmr.2014.2517