LSD1-mediated epigenetic modification contributes to ovarian cancer cell migration and invasion

  • Authors:
    • Yuanxia Li
    • Xiaolei Wan
    • Ye Wei
    • Xiuwen Liu
    • Wensheng Lai
    • Liuping Zhang
    • Jie Jin
    • Chaoyang Wu
    • Qixiang Shao
    • Genbao Shao
    • Qiong Lin
  • View Affiliations

  • Published online on: April 4, 2016     https://doi.org/10.3892/or.2016.4729
  • Pages: 3586-3592
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lysine-specific demethylase 1 (LSD1) has been implicated in the process of tumor progression at various steps, but its role in epithelial-messenchymal transition (EMT) and the migration of ovarian cancer cells remains obscure. In this study, we demonstrated the effect of LSD1 on ovarian cancer cell migration and the regulatory role of LSD1 in the expression of EMT markers. Inhibition of LSD1 expression impaired the migration and invasion of HO8910 ovarian cancer cells. In contrast, overexpression of LSD1 enhanced the cell migration and invasion of HO8910 cells. Mechanistic analyses showed that LSD1 promoted cell migration through induction of N-cadherin, vimentin, MMP-2 and inhibition of E-cadherin. Furthermore, LSD1 interacted with the promoter of E-cadherin and demethylated histone H3 lysine 4 (H3K4) at this region, downregulated E-cadherin expression, and consequently enhanced ovarian cancer cell migration. These data indicate that LSD1 acts as an epigenetic regulator of EMT and contributes to the metastasis of ovarian cancer.

Introduction

Ovarian cancer is the second most common cancer among female gynecologic cancers and has become the leading cause of cancer-related death among females (1). Due to the difficulty in early detection, 75% of ovarian cancer patients are diagnosed at advanced stages (stage III or IV) (2). In stage III or IV, the tumor involves one or both ovaries with peritoneal metastasis outside the pelvis or distant metastasis to liver parenchyma or other visceral organs (2,3). Early invasion and metastasis have been well accepted as the leading features and main causes of death in ovarian cancer. However, mechanistic understanding of the metastatic potential of ovarian cancer remains unclear, and novel targets are yet to be identified for treating metastatic ovarian cancer.

Lysine-specific demethylase 1 (LSD1/KDM1A/AOF2) is the first histone demethylase discovered, which specifically demethylates mono- and dimethylated histone H3 lysine 4 (H3K4) and histone H3 lysine 9 (H3K9) (4). LSD1 is frequently overexpressed in lung cancer (5,6), breast cancer (7), prostate cancer (8,9), and liver cancer (10). Importantly, overexpression of LSD1 promotes the growth and invasion of various types of cancer cells, and contributes to human carcinogenesis by regulating the expression of genes involved in various chromatin-modifying pathways (6). Conversely, inhibition of LSD1 was found to suppress cell invasion and migration in various types of cancers (5,11,12). Although LSD1 is recently described to be highly expressed in ovarian cancer (13,14), the biological function of LSD1 in this cancer remains largely unknown.

Epithelial-messenchymal transition (EMT) is a process whereby epithelial cells are programmed into mesenchymal cells (15). EMT is now considered as the initial and essential step in tumor metastasis. During EMT, epithelial cells acquire cell motility by reducing cell-cell junctions, and loss of cell polarity (16,17). E-cadherin, an epithelial marker, has a crucial role in regulating cell-cell adhesion and maintenance of tissue architecture (18). Indeed, E-cadherin serves as a suppressor of cell migration and invasion (1922). Transcription factors, including Snail, Slug, Zeb1 and Twist, can induce EMT by downregulating E-cadherin expression (2326). Recent studies show that LSD1 is recruited by the transcription factor Snail to the promoter of E-cadherin to repress the expression of the E-cadherin gene consequently contributing to cancer cell invasion (27,28). Conversely, Ferrari-Amorotti et al observed that blocking Snail-LSD1 interaction by treatment with Parnate suppressed the invasiveness of cancer cells (29).

Few studies have reported on how LSD1 induces EMT and finally contributes to ovarian cancer cell migration. Therefore in the present study, we examined the effect of LSD1 on cell migration and invasion using LSD1-knockdown and overexpressing HO8910 ovarian cancer cells as models. We also examined the regulatory role of LSD1 in the expression of molecular markers of EMT. Knockdown of LSD1 reduced cell migration and invasion in the HO8910 cells, while overexpression of LSD1 stimulated the migration and invasion of the HO8910 cells. Mechanistic analyses uncovered that LSD1 promoted cell migration through induction of N-cadherin, Snail, vimentin, MMP-2 and inhibition of E-cadherin. LSD1 epigenetically regulated the transcription of E-cadherin through demethylating H3K4 at the E-cadherin promoter. Collectively, these results suggest that targeting LSD1 may be a novel therapeutic approach for the treatment of ovarian cancer.

Materials and methods

Cell lines and cell culture

The human ovarian cancer cell line, HO8910, was kindly provided by Dr Qixiang Shao of Jiangsu University (Zhenjiang, China). HO8910 cells were cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum (FBS) (both from Gibco, Grand Island, NY, USA) at a temperature of 37°C under 5% CO2. HEK 293T cells were cultured in Dulbecco's modified Eagle's medium (DMEM; Gibco) containing 10% FBS at a temperature of 37°C under 5% CO2.

Antibodies and reagents

The pLKO-Tet-On, pLVX-tight-puro, pHR′-CMV-8.2ΔVPR, and pHR′-CMV-VSVG vectors were kind gifts from Dr Changdeng Hu (Purdue University, West Lafayette, in, USA). LSD1, E-cadherin, Snail, vimentin, N-cadherin and MMP-2 antibodies were purchased from Cell Signaling Technology Inc. (Danvers, MA, USA). The α-tubulin and horseradish peroxidase (HRP)-conjugated goat anti-rabbit antibodies were obtained from Bioworld Technology (Shanghai, China). Electrochemiluminescence (ECL) reagents were purchased from Millipore Corp. (Billerica, MA, USA). H3K4me2 antibody was purchased from upstate biotechnology Inc. (Lake Placid, NY, USA). Polybrene, doxycycline (Dox), puromycin and G418 were purchased from Sigma-Aldrich (St. Louis, MO, USA). The LSD1 inhibitor tranylcypromine (TCP) was obtained from Biomol International (Plymouth Meeting, PA, USA).

Plasmid constructions and transfections

For generation of the shRNA-LSD1 plasmid, annealed short hairpin oligonucleotides (the RNAi Consortium collection TRCN0000046072; Sigma-Aldrich) targeting CCACGAGTCAAACCTTTATTT in the coding regions (CDS) of LSD1 were cloned into pLKO-Tet-On by AgeI and EcoRI sites to produce pLKO-Tet-On-shLSD1 as described previously (30,31). The constructs were confirmed by DNA sequencing. All transfections were performed using the Lipofectamine 2000 reagent (Invitrogen) according to the manufacturer's instructions.

Establishment of the stable cell lines (LSD1-knockdown and overexpressing)

To generate lentiviral particles, 293T cells were seeded in 6-cm dishes and transfected with 2 µg of pLKO-Tet-On-shLSD1, 1.5 µg of pHR′-CMV-8.2ΔVPR and 0.5 µg of pHR′-CMV-VSVG using Lipofectamine 2000 reagent. The supernatant containing the lentiviral particles was harvested 24, 48 and 72 h post-transfection, and then centrifuged (124 × g for 5 min) to remove cell debris. HO8910 cells cultured in 6-cm dishes were infected by adding 1 ml lentiviral supernatant and 3 ml complete medium containing 8 µg/ml Polybrene. After the infection (twice), cells were selected with 2.0 µg/ml puromycin for 3 days and then maintained with 1.0 µg/ml puromycin for one week.

To generate rTet-repressor expressing (rtTA) cell line, 293T cells were transfected with 2 µg of pLVX-Tet-On, 1.5 µg of pHR′-CMV-8.2ΔVPR and 0.5 µg of pHR′-CMV-VSVG using Lipofectamine 2000 reagent. After transfection (24 h), the viral supernatant was harvested and used to infect HO8910 cells. After the infection (twice), HO8910 cells were selected with 200 µg/ml G418 for 1 week. The cells that survived were stable rtTA. HO8910-rtTA cells were then infected with the lentiviral particles packaged with pLVX-tight-puro-LSD1. After infection twice, HO8910-rtTA cells were selected with 2.0 µg/ml of puromycin for 3 days, and then maintained in the presence of 1.0 µg/ml of puromycin for one week. The surviving cells were considered as stable clones. The stable clones were further confirmed by western blot analysis.

RNA extraction and real-time RT-PCR (qRT-PCR)

Total RNA was isolated from the cells using RNAiso plus (Takara, Shiga, Japan) and reverse-transcribed using the PrimeScript RT reagent kit (Takara) to generate cDNAs. Then the cDNAs were subjected to qRT-PCR as described previously (32). qRT-PCR was performed with SYBR-Green PCR Master Mix (Takara) on a Bio-Rad CFX96 system (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The primer sequences used were: LSD1 (GenBank accession no. NM 015013.3), 5′-CAAGTGTCAATTTGTTCGGG-3′ (forward) and 5′-TTCTTTGGGCTGAGGTACTG-3′ (reverse); and GAPDH (GenBank accession no. NM001256799.1), 5′-GCAAATTCCATGGCACCGTC-3′ (forward) and 5′-TCGCCCCACTTGATTTTGG-3′ (reverse). The relative quantification of mRNA levels was normalized to levels of GADPH and calculated by comparative 2−ΔΔCt.

Western blot analysis

Protein lysates were extracted from the cells and blotted as described previously (33). Equal amounts of soluble proteins were electrophoresed by SDS-PAGE and transferred to 0.45-µm PVDF membranes. The membranes were blocked with 5% nonfat-dry milk for 1 h at room temperature (RT). After incubation with the primary antibodies against LSD1 (1:1,000), E-cadherin (1:500), Snail (1:500), vimentin (1:500), N-cadherin (1:500) or MMP-2 (1:500) overnight at 4°C and with the corresponding secondary antibodies (1:5,000) for 1 h at RT, the immunoblots were developed by ECL method.

Migration and invasion assays

For the invasion assay, each Boyden chamber (BD Biosciences, Bedford, MA, USA) was coated with 60 µl Matrigel diluted with DMEM (1:30) and incubated at 37°C for 4–6 h. Cells (1.5×105) were resuspended with DMEM containing Dox or TCP in the upper chamber. Then, 10% FBS-containing medium was placed in the lower chamber to act as a chemoattractant. After a 24-h incubation, the non-invading cells remaining on the upper surface were removed, and the cells on the lower surface were fixed with 4% formaldehyde for 30 min, and stained with 0.1% crystal violet for 15 min. At least 5 fields for each chamber were photographed (×200 magnification) and counted, and the invading cells were counted in each field. The cell migration assay was performed using Boyden chambers without Matrigel coating. All experiments were performed at least in triplicate.

Chromatin immunoprecipitation (ChIP)

All reagents were provided by Upstate Biotechnology (EZ-ChIP™ kit 17-371). Cells were fixed with 1% formaldehyde to cross-link proteins. The reaction was stopped by adding 10X glycine. Cross-linked cells were washed with PBS twice, pelleted and resuspended in SDS lysis buffer at a concentration of 1×107 cells/ml. Aliquots of 400 µl were sonicated with 4–6 sets of 5-sec pulses (32% output) on ice. Then sonicated lysates were centrifuged and divided into 100 µl aliquots for each ChIP assay (1×106 cells/IP), and precleared with protein G-agarose. After incubation with the antibodies overnight at 4°C, immune complexes were collected with protein G-agarose, and then washed with low salt immune complex wash buffer, high salt immune complex wash buffer, and finally TE buffer. The immune complexes were eluted with 20% SDS, and 1 M NaHCO3. The crosslinks were reversed overnight at 65°C, then the DNA was purified using spin columns, and finally subjected to qRT-PCR. Chromatin eluted from the IPs with IgG and anti-RNA polymerase were used as the negative and positive control, respectively. Two previously described primers of E-cadherin promoter for ChIP (34,35) were as follows: E-ca01 5′-GGGCAATACAGGGAGACACA-3′ (forward) and 5′-GGGCTTTTACACTTGGCTGA-3′ (reverse); E-ca02 5′-CACAACAGCATAGGGAGACATT-3′ (forward) and 5′-TGTAGAGCTTCATGGGTTAGTGA-3′ (reverse).

Statistical analysis

All values are presented as the mean ± SEM. The data were analyzed using the Student's t-test with SPSS 11.5 software (SPSS Inc.). P-values with a 95% confidence interval were obtained from at least three independent experiments. A p-value <0.01 was considered to indicate a statistically significant result.

Results

LSD1 is required for cell migration and invasion in ovarian cancer cells

To investigate the contribution of LSD1 to the migration and invasion of ovarian cancer HO8910 cells, we generated stable LSD1-knockdown (LSD1-KD) clones and LSD1-overexpressing (LSD1-OE) clones from the HO8910 cells. Total RNA and proteins were extracted from these stable cells treated with increasing doses of Dox for 24 or 48 h. Our results showed the mRNA and protein expression of the LSD1 gene was decreased in the LSD1-KD cells in a dose-dependent manner (Fig. 1A and B), whereas the levels of LSD1 mRNA and protein expression were increased in the LSD1-OE cells (Fig. 1C–E).

To understand the effect of LSD1 expression on cell migration and invasion, we performed Transwell assays to measure the migratory capacity of these two transfected cell lines. The LSD1-KD cells displayed less migration and invasion in comparison with the control (Fig. 2A and B), whereas the LSD1-OE cells had a higher rate of migration and invasion as compared to the control (Fig. 2C and D).

To further determine the role of LSD1 in cell migration, we utilized a known potent inhibitor, TCP (30,36), to suppress the demethylase activity of LSD1 in HO8910 cells. Inhibition of LSD1 decreased the migration activity of the HO8910 cells in a dose-dependent manner (Fig. 3A and B). Taken together, these data suggest that LSD1 is essential for cell migration and invasion in HO8910 ovarian cancer cells.

LSD1 regulates EMT in ovarian cancer cells

As epithelial-mesenchymal transition (EMT) is involved in tumor migration and invasion, we examined the expression of several EMT markers in the LSD1-KD and LSD1-OE HO8910 cells. We found that knockdown of LSD1 upregulated the expression of the epithelial marker E-cadherin and downregulated the expression of the mesenchymal markers N-cadherin, vimentin and MMP-2 (Fig. 4A). LSD1 knockdown also caused a decrease in the expression of the transcription factor Snail (Fig. 4A). Furthermore, inhibition of LSD1 induced an increase in E-cadherin expression and a decrease in the expression of N-cadherin, vimentin, MMP-2 and Snail in a dose-dependent manner (Fig. 4C). On the contrary, overexpression of LSD1 induced a decrease in E-cadherin expression, with a concomitant increase in the expression of N-cadherin, Vimentin, MMP-2 and Snail in the HO8910 cells (Fig. 4B).

LSD1 knockdown increases H3K4me2 levels at the E-cadherin promoter

Given that knockdown of LSD1 was accompanied by the upregulation of E-cadherin at the transcriptional level (Fig. 5A) and inhibition of migration of ovarian cancer cells (Fig. 2A and B), we speculated that LSD1 could enhance migration by downregulating E-cadherin expression via demethylation of H3K4me2, a major substrate of LSD1 in ovarian cancer cells (30). To confirm this speculation, Chip assays were performed in the LSD1-KD HO8910 cells incubated with the anti-H3K4me2 antibody. Quantitative analysis indicated that the enrichment of H3K4me2 at the promoter of the e-cadherin gene was significantly higher in the LSD1-KD cells than that in the control cells (Fig. 5B). Collectively, our data revealed that the expression of LSD1 caused a decrease in H3K4me2 levels at the E-cadherin promoter, reduced E-cadherin expression, and consequently contributed to the migration of HO8910 cells.

Discussion

Ovarian cancer is the second most common malignant gynecologic tumor, and represents the leading cause of cancer-related death among women worldwide (1). The high mortality rate of ovarian cancer is caused by tumor metastasis, post-surgical recurrence, and late detection at advanced stages (3). Ovarian cancer is associated with multiple risk factors and is currently recognized as both a genetic and epigenetic disease (37,38). While the genetic changes in ovarian cancer have been extensively studied, the contribution of epigenetic alterations to ovarian cancer progression remains poorly understood. Histone methylation is a dynamic epigenetic process that has been found to be associated with cancer, including ovarian cancer (39). LSD1 is a well-characterized demethylase that can remove methyl groups from H3K4 (4). However, its role and underlying mechanisms in ovarian cancer are still unclear. In this study, we showed that LSD1 overexpression induced EMT, migration and invasion of HO8910 ovarian cancer cells. In contrast, silencing of LSD1 reversed these events in invasive HO8910 cells. We also showed a mechanistic link between LSD1 and E-cadherin through LSD1-mediated regulation of H3K4me2, which subsequently leads to the downregulation of E-cadherin transcription.

Histone demethylases are epigenetic enzymes that can remove both repressive and activating histone marks. LSD1 family members are capable of removing the H3K4me2-activating marks and rendering them potential players in the downregulation of tumor suppressors (40,41). The putative role of LSD1 as an oncogene in cancer development is supported by the observation that LSD1 is highly expressed in ovarian cancer (13,14) and other malignant tumors (510). LSD1 is reported to play an important role in ovarian cancer cell proliferation via a Sox2-mediated mechanism (31). Our present study points to a novel function of LSD1 in ovarian cancer cell migration and invasion through regulation of EMT.

Recently, the regulation of epigenetic modification on EMT is a hot topic. Several studies have shown that histone modifications are involved in Snail-mediated transcriptional repression of E-cadherin. Peinado et al reported that Snail induces repressive histone modifications at the E-cadherin promoter through recruitment of histone deacetylases (HDACs) (42). Recent studies have demonstrated that Snail recruits LSD1 to the E-cadherin promoter to reduce E-cadherin expression by removing H3K4me2 (27,28). In this study, we found that modulation of LSD1 expression alters the methylation status of H3K4 at the E-cadherin promoter, which in turn transcriptionally regulates the expression of E-cadherin. Thus, we conclude that LSD1 transcriptionally downregulates E-cadherin expression via H3K4 demethylation, and consequently results in the increased migration and invasion of HO8910 cells.

Taking all these pieces of evidence together, we are able to show that knockdown of LSD1 impairs the migration and invasion of HO8910 cells by regulating EMT, while overexpression of LSD1 has a converse effect on cell migration. By demethylating H3K4me2 at the E-cadherin promoter, LSD1 downregulates the E-cadherin expression, and contributes to the metastasis of HO8910 cells. Our results suggest that LSD1 may be a potential therapeutic target for metastatic ovarian cancer.

Acknowledgments

This study was supported by grants from the National Natural Science Foundation of China (81170573) and Clinical Medicine Science & Technology Project of Jiangsu Province of China (BL2013024).

References

1 

Jemal A, Siegel R, Xu J and Ward E: Cancer statistics, 2010. CA Cancer J Clin. 60:277–300. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Hennessy BT, Coleman RL and Markman M: Ovarian cancer. Lancet. 374:1371–1382. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Chaffer CL and Weinberg RA: A perspective on cancer cell metastasis. Science. 331:1559–1564. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Shi Y, Lan F, Matson C, Mulligan P, Whetstine JR, Cole PA, Casero RA and Shi Y: Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell. 119:941–953. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Lv T, Yuan D, Miao X, Lv Y, Zhan P, Shen X and Song Y: Overexpression of LSD1 promotes proliferation, migration and invasion in non-small cell lung cancer. PLoS One. 7:e350652012. View Article : Google Scholar

6 

Hayami S, Kelly JD, Cho HS, Yoshimatsu M, Unoki M, Tsunoda T, Field HI, Neal DE, Yamaue H, Ponder BA, et al: Overexpression of LSD1 contributes to human carcinogenesis through chromatin regulation in various cancers. Int J Cancer. 128:574–586. 2011. View Article : Google Scholar

7 

Serce N, Gnatzy A, Steiner S, Lorenzen H, Kirfel J and Buettner R: Elevated expression of LSD1 (Lysine-specific demethylase 1) during tumour progression from pre-invasive to invasive ductal carcinoma of the breast. BMC Clin Pathol. 12:132012. View Article : Google Scholar : PubMed/NCBI

8 

Wu CY, Hsieh CY, Huang KE, Chang C and Kang HY: Cryptotanshinone down-regulates androgen receptor signaling by modulating lysine-specific demethylase 1 function. Int J Cancer. 131:1423–1434. 2012. View Article : Google Scholar

9 

Kahl P, Gullotti L, Heukamp LC, Wolf S, Friedrichs N, Vorreuther R, Solleder G, Bastian PJ, Ellinger J, Metzger E, et al: Androgen receptor coactivators lysine-specific histone demethylase 1 and four and a half LIM domain protein 2 predict risk of prostate cancer recurrence. Cancer Res. 66:11341–11347. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Zhao Z-K, Yu H-F, Wang D-R, Dong P, Chen L, Wu WG, Ding WJ and Liu YB: Overexpression of lysine specific demethylase 1 predicts worse prognosis in primary hepatocellular carcinoma patients. World J Gastroenterol. 18:6651–6656. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Ding J, Zhang ZM, Xia Y, Liao GQ, Pan Y, Liu S, Zhang Y and Yan ZS: LSD1-mediated epigenetic modification contributes to proliferation and metastasis of colon cancer. Br J Cancer. 109:994–1003. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Yu Y, Wang B, Zhang K, Lei Z, Guo Y, Xiao H, Wang J, Fan L, Lan C, Wei Y, et al: High expression of lysine-specific demethylase 1 correlates with poor prognosis of patients with esophageal squamous cell carcinoma. Biochem Biophys Res Commun. 437:192–198. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Konovalov S and Garcia-Bassets I: Analysis of the levels of lysine-specific demethylase 1 (LSD1) mRNA in human ovarian tumors and the effects of chemical LSD1 inhibitors in ovarian cancer cell lines. J Ovarian Res. 6:752013. View Article : Google Scholar : PubMed/NCBI

14 

Chen C, Ge J, Lu Q, Ping G, Yang C and Fang X: Expression of lysine-specific demethylase 1 in human epithelial ovarian cancer. J Ovarian Res. 8:282015. View Article : Google Scholar : PubMed/NCBI

15 

Hay ED: An overview of epithelio-mesenchymal transformation. Acta Anat (Basel). 154:8–20. 1995. View Article : Google Scholar

16 

Boyer B, Vallés AM and Edme N: Induction and regulation of epithelial-mesenchymal transitions. Biochem Pharmacol. 60:1091–1099. 2000. View Article : Google Scholar : PubMed/NCBI

17 

Thiery JP: Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2:442–454. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Angst BD, Marcozzi C and Magee AI: The cadherin superfamily. J Cell Sci. 114:625–626. 2001.PubMed/NCBI

19 

Hazan RB, Qiao R, Keren R, Badano I and Suyama K: Cadherin switch in tumor progression. Ann NY Acad Sci. 1014:155–163. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Perl AK, Wilgenbus P, Dahl U, Semb H and Christofori G: A causal role for E-cadherin in the transition from adenoma to carcinoma. Nature. 392:190–193. 1998. View Article : Google Scholar : PubMed/NCBI

21 

Birchmeier W, Hulsken J and Behrens J: E-cadherin as an invasion suppressor. Ciba Foundation Symposium. 189:124–136; discussion 136–141, 174–126. 1995.PubMed/NCBI

22 

Batlle E, Sancho E, Francí C, Domínguez D, Monfar M, Baulida J and García De Herreros A: The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nat Cell Biol. 2:84–89. 2000. View Article : Google Scholar : PubMed/NCBI

23 

Kang Y and Massagué J: Epithelial-mesenchymal transitions: twist in development and metastasis. Cell. 118:277–279. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Kato Y, Yashiro M, Noda S, Tendo M, Kashiwagi S, Doi Y, Nishii T, Matsuoka J, Fuyuhiro Y, Shinto O, et al: Establishment and characterization of a new hypoxia-resistant cancer cell line, OCUM-12/Hypo, derived from a scirrhous gastric carcinoma. Br J Cancer. 102:898–907. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA, Come C, Savagner P, Gitelman I, Richardson A and Weinberg RA: Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell. 117:927–939. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Waldmann J, Feldmann G, Slater EP, Langer P, Buchholz M, Ramaswamy A, Saeger W, Rothmund M and Fendrich V: Expression of the zinc-finger transcription factor Snail in adrenocortical carcinoma is associated with decreased survival. Br J Cancer. 99:1900–1907. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Lin Y, Wu Y, Li J, Dong C, Ye X, Chi YI, Evers BM and Zhou BP: The SNAG domain of Snail1 functions as a molecular hook for recruiting lysine-specific demethylase 1. EMBO J. 29:1803–1816. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Lin T, Ponn A, Hu X, Law BK and Lu J: Requirement of the histone demethylase LSD1 in Snai1-mediated transcriptional repression during epithelial-mesenchymal transition. Oncogene. 29:4896–4904. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Ferrari-Amorotti G, Fragliasso V, Esteki R, Prudente Z, Soliera AR, Cattelani S, Manzotti G, Grisendi G, Dominici M, Pieraccioli M, et al: Inhibiting interactions of lysine demethylase LSD1 with snail/slug blocks cancer cell invasion. Cancer Res. 73:235–245. 2013. View Article : Google Scholar :

30 

Shao G, Wang J, Li Y, Liu X, Xie X, Wan X, Yan M, Jin J, Lin Q, Zhu H, et al: Lysine-specific demethylase 1 mediates epidermal growth factor signaling to promote cell migration in ovarian cancer cells. Sci Rep. 5:153442015. View Article : Google Scholar : PubMed/NCBI

31 

Zhang X, Lu F, Wang J, Yin F, Xu Z, Qi D, Wu X, Cao Y, Liang W, Liu Y, et al: Pluripotent stem cell protein Sox2 confers sensitivity to LSD1 inhibition in cancer cells. Cell Reports. 5:445–457. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Shao GB, Wang J, Zhang LP, Wu CY, Jin J, Sang JR, Lu HY, Gong AH, Du FY and Peng WX: Aging alters histone H3 lysine 4 methylation in mouse germinal vesicle stage oocytes. Reprod Fertil Dev. 27:419–426. 2015. View Article : Google Scholar

33 

Zhang L, Wang J, Pan Y, Jin J, Sang J, Huang P and Shao G: Expression of histone H3 lysine 4 methylation and its demethylases in the developing mouse testis. Cell Tissue Res. 358:875–883. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Li LC, Okino ST, Zhao H, Pookot D, Place RF, Urakami S, Enokida H and Dahiya R: Small dsRNAs induce transcriptional activation in human cells. Proc Natl Acad Sci USA. 103:17337–17342. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Ting AH, Schuebel KE, Herman JG and Baylin SB: Short double-stranded RNA induces transcriptional gene silencing in human cancer cells in the absence of DNA methylation. Nat Genet. 37:906–910. 2005. View Article : Google Scholar : PubMed/NCBI

36 

Sun G, Alzayady K, Stewart R, Ye P, Yang S, Li W and Shi Y: Histone demethylase LSD1 regulates neural stem cell proliferation. Mol Cell Biol. 30:1997–2005. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Verma M, Seminara D, Arena FJ, John C, Iwamoto K and Hartmuller V: Genetic and epigenetic biomarkers in cancer: improving diagnosis, risk assessment, and disease stratification. Mol Diagn Ther. 10:1–15. 2006. View Article : Google Scholar

38 

Seeber LM and van Diest PJ: Epigenetics in ovarian cancer. Methods Mol Biol. 863:253–269. 2012. View Article : Google Scholar : PubMed/NCBI

39 

He Y, Korboukh I, Jin J and Huang J: Targeting protein lysine methylation and demethylation in cancers. Acta Biochim Biophys Sin (Shanghai). 44:70–79. 2012. View Article : Google Scholar

40 

Chen Y, Jie W, Yan W, Zhou K and Xiao Y: Lysine-specific histone demethylase 1 (LSD1): a potential molecular target for tumor therapy. Crit Rev Eukaryot Gene Expr. 22:53–59. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Lim S, Janzer A, Becker A, Zimmer A, Schüle R, Buettner R and Kirfel J: Lysine-specific demethylase 1 (LSD1) is highly expressed in ER-negative breast cancers and a biomarker predicting aggressive biology. Carcinogenesis. 31:512–520. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Peinado H, Ballestar E, Esteller M and Cano A: Snail mediates E-cadherin repression by the recruitment of the Sin3A/histone deacetylase 1 (HDAC1)/HDAC2 complex. Mol Cell Biol. 24:306–319. 2004. View Article : Google Scholar :

Related Articles

Journal Cover

June-2016
Volume 35 Issue 6

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li Y, Wan X, Wei Y, Liu X, Lai W, Zhang L, Jin J, Wu C, Shao Q, Shao G, Shao G, et al: LSD1-mediated epigenetic modification contributes to ovarian cancer cell migration and invasion. Oncol Rep 35: 3586-3592, 2016
APA
Li, Y., Wan, X., Wei, Y., Liu, X., Lai, W., Zhang, L. ... Lin, Q. (2016). LSD1-mediated epigenetic modification contributes to ovarian cancer cell migration and invasion. Oncology Reports, 35, 3586-3592. https://doi.org/10.3892/or.2016.4729
MLA
Li, Y., Wan, X., Wei, Y., Liu, X., Lai, W., Zhang, L., Jin, J., Wu, C., Shao, Q., Shao, G., Lin, Q."LSD1-mediated epigenetic modification contributes to ovarian cancer cell migration and invasion". Oncology Reports 35.6 (2016): 3586-3592.
Chicago
Li, Y., Wan, X., Wei, Y., Liu, X., Lai, W., Zhang, L., Jin, J., Wu, C., Shao, Q., Shao, G., Lin, Q."LSD1-mediated epigenetic modification contributes to ovarian cancer cell migration and invasion". Oncology Reports 35, no. 6 (2016): 3586-3592. https://doi.org/10.3892/or.2016.4729