Gemcitabine reactivates epigenetically silenced genes and functions as a DNA methyltransferase inhibitor

  • Authors:
    • Steven G. Gray
    • Anne-Marie Baird
    • Fardod O'Kelly
    • Georgios Nikolaidis
    • Malin Almgren
    • Armelle Meunier
    • Eilis Dockry
    • Donal Hollywood
    • Tomas J. Ekström
    • Antoinette S. Perry
    • Kenneth J. O'Byrne
  • View Affiliations

  • Published online on: September 21, 2012     https://doi.org/10.3892/ijmm.2012.1138
  • Pages: 1505-1511
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gemcitabine is indicated in combination with cisplatin as first-line therapy for solid tumours including non-small cell lung cancer (NSCLC), bladder cancer and mesothelioma. Gemcitabine is an analogue of pyrimidine cytosine and functions as an anti-metabolite. Structurally, however, gemcitabine has similarities to 5-aza-2-deoxycytidine (decitabine/Dacogen®), a DNA methyltransferase inhibitor (DNMTi). NSCLC, mesothelioma and prostate cancer cell lines were treated with decitabine and gemcitabine. Reactivation of epigenetically silenced genes was examined by RT-PCR/qPCR. DNA methyltransferase activity in nuclear extracts and recombinant proteins was measured using a DNA methyl­transferase assay, and alterations in DNA methylation status were examined using methylation-specific PCR (MS-PCR) and pyrosequencing. We observe a reactivation of several epigenetically silenced genes including GSTP1, IGFBP3 and RASSF1A. Gemcitabine functionally inhibited DNA methyltransferase activity in both nuclear extracts and recombinant proteins. Gemcitabine dramatically destabilised DNMT1 protein. However, DNA CpG methylation was for the most part unaffected by gemcitabine. In conclusion, gemcitabine both inhibits and destabilises DNA methyltransferases and reactivates epigenetically silenced genes having activity equivalent to decitabine at concentrations significantly lower than those achieved in the treatment of patients with solid tumours. This property may contribute to the anticancer activity of gemcitabine.

Introduction

Gemcitabine is a well-established anticancer agent and has been FDA approved either as a single agent for the treatment of pancreatic cancer (1) or in combination with another agent (carboplatin, cisplatin or paclitaxel) for the treatment of ovarian (2) and metastatic breast cancer (3). Cisplatin is indicated as first-line therapy for inoperable, locally advanced (stage IIIA or IIIB) or metastatic (stage IV) non-small cell lung cancer (NSCLC) (4,5), and has also been studied in other solid tumours such as malignant pleural mesothelioma (MPM) (6,7) and prostate cancer (8,9).

Gemcitabine is considered to act as a pyrimidine-type anti-metabolite. The agent is a prodrug and requires intracellular conversion to two active metabolites, gemcitabine diphosphate and gemcitabine triphosphate, which can function in two ways: by binding to and irreversibly inhibiting ribonucleotide reductase (RNR) and by replacing one of the building blocks of nucleic acids, in this case cytosine, during DNA replication (10).

Aberrant epigenetic regulation of gene expression is a frequent event in cancer (11). Several of the proteins which regulate histone post-translational modifications have now been shown to play a role in resistance to various cancer therapies including cisplatin (12), gefitinib (13), etoposide (14) and tamoxifen (15).

DNA CpG methylation is another epigenetic modification that is linked to loss of gene expression in cancer (16). Inhibitors targeting DNA methyltransferases have been developed. Of these 5-azacytidine (Vidaza®) and 5-aza-2-deoxycytidine (decitabine/Dacogen®) have gained FDA approval for the treatment of myelodysplastic syndrome subtypes (17,18).

Notably, the chemotherapy agent doxorubicin has been shown to inhibit the DNA methyltransferase DNMT1 (19) and indicates that other chemotherapy drugs may potentially inhibit the epigenetic enzymatic machinery. Since decitabine and gemcitabine are cytosine analogues, and share similar structural features (Fig. 1), we reasoned that gemcitabine may also be able to act as a DNA methyltransferase inhibitor (DNMTi).

Materials and methods

Cell lines

The A549 (adenocarcinoma), SKMES1 (squamous cell carcinoma), H-28 (MPM), 22Rv1 (prostate), LNCaP (prostate) and Du145 (prostate) cell lines were purchased from the ATCC (LGC Promochem). Cells were cultured in appropriate media and maintained at 37°C in a humidified atmosphere containing 5% CO2.

Cell line treatments

Decitabine was purchased from Merck and dissolved in methanol. Cell cultures were treated for 48 h at a final concentration of 5 μM, with the media and drug replaced every 24 h as previously described (20).

Gemcitabine was supplied by Eli Lilly and dissolved in phosphate-buffered saline (PBS) at a final concentration of 38 mg/ml (120.14 mM). Cell cultures were treated for 48 h at final concentrations of 0.2, 0.5 or 1 μM with the media and drug replaced every 24 h. We chose to use this concentration based on literature searches in Pubmed.

RNA isolation and RT-PCR amplification

Total RNA was extracted using TRI Reagent® (MRCgene) according to the manufacturer’s instructions. One microgram of total RNA was used to generate cDNA using M-MLV-reverse transcriptase (Promega) according to the manufacturer’s instructions.

Expression of VEGFR1, VEGFR2, sFRP4, RASSF1A and β-actin was examined by RT-PCR, using primers and PCR conditions outlined in Table I. Each PCR was carried out for 35 cycles. Ten microlitres of the experimental RT-PCR product and 2 μl of the β-actin RT-PCR product were loaded onto a 1% agarose gel.

Table I

Primers and annealing temperatures for RT-PCR.

Table I

Primers and annealing temperatures for RT-PCR.

Primer SetSequenceAnnealing Temp (°C)/PCR cycles
VEGFR1F: 5′-CAAGTGGCCAGAGGCATGGAGTT-3′56°C/35 cycles
R:5′-GATGTAGTCTTTACCATCCTGTTG-3′
VEGFR2F: 5′-GAGGGCCTCTCATGGTGATTGT-3′56°C/35 cycles
R: 5′-TGCCAGCAGTCCAGCATGGTCTG-3′
sFRP4F: 5′-TCTATGACCGTGGCGTGTGC-3′56°C/35 cycles
R: 5′-ACCGATCGGGGCTTAGGCGTTTAC-3′
RASSF1AF: 5′-CAGATTGCAAGTTCACCTGCCACTA-3′56°C/35 cycles
R: 5′-GATGAAGCCTGTGTAAGAACCGTCCT
β-actinF: 5′-TGTTTGAGACCTTCAACACCC-3′56°C/35 cycles
R: 5′-AGCACTGTGTTGGCGTACAG-3′

[i] F, forward; R, reverse.

Quantitative PCR

RNA was isolated from the cell lines using a miRVana miRNA isolation kit (Applied Biosystems, UK) according to the manufacturer’s guidelines. Total RNA (1 μg) was reverse transcribed using the high capacity cDNA Archive kit. Expression of GSTP1 (TaqMan® gene expression assay ID: Hs00168310_m1) and IGFBP3 (TaqMan gene expression assay ID: Hs00181211_m1) was quantified in the cell lines by qRT-PCR. Human phosphoglycerate kinase 1 (PGK1) was used as an endogenous control (TaqMan gene expression assay ID: Hs99999906_m1). TaqMan PCR reactions were performed in triplicate on an ABI Prism 7900 sequence detection system. Gene expression was calculated relative to the untreated cell lines, using SDS RQ Manager 1.2 software, which automatically determines relative quantities (RQ), by applying the arithmetic formula 2−ΔΔCT. All equipment and reagents were supplied by Applied Biosystems, Foster City, CA.

DNA methyltransferase assay

Analysis of DNA methyltransferase activity was carried out using the EpiQuik™ DNA methyltransferase activity/inhibition assay kit (Epigentek) using both recombinant DNMTs (Epigentek) and nuclear extracts isolated with the EpiQuik Nuclear Extraction kit II (Epigentek) according to the manufacturer’s instructions. Following consultation with the manufacturers, the recombinant proteins were incubated with the various concentrations of decitabine or gemcitabine for 90 min at room temperature prior to conducting the methyltransferase assay.

Genomic DNA isolation, bisulfite conversion and MS-PCR analysis

Genomic DNA was isolated from the cell lines using a solution containing 0.5% SDS and 100 μg/ml Proteinase K (21).

Genomic DNA from the cell lines (500 ng) was bisulfite modified using the EpiTect Bisulfite kit (Qiagen, UK), following the manufacturer’s guidelines. EpiTect methylated DNA and unmethylated DNA were used as controls.

Promoter hypermethylation of the GSTP1 promoter was analysed by methylation-specific PCR (MS-PCR) in the LNCaP and 22Rv1 cell lines using the GoTaq HotStart enzyme (Promega). PCR primer sets complementary to both modified, methylated DNA (M) and modified, unmethylated DNA (U) were designed for GSTP1: GSTP1 MF2, 5′-TTCGGGGGTGTA GCGGTCGTC-3′; GSTP1 MR1, 5′-CCAACGAAAACCTCGC GACCTCCG-3′ (expected product, 145 bp); GSTP1 UF2, 5′-GATGTTTGGGGTGTAGTGGTTGTT-3′; GSTP1 UR1, 5′-AAACTCCAACAAAAACCTCACAACCTCCA-3′ (expected product, 154 bp).

Bisulfite pyrosequencing

Pyrosequencing methylation analysis (PMA) of the LINE-1.2 element, the RASSF1A promoter and the VEGFR gene promoters, was performed as previously described (2224). Genomic DNA (500–1,000 ng) was bisulfite treated, using the EpiTect Bisulfite kit or the EZ DNA Methylation™ kit (ZymoResearch). The PyroMarkAssay Design software was used to design the primers for amplification and sequencing to cover a number of CpG sites as shown in Table II. PCR amplification products were cleaned and subjected to pyrosequencing on either a PyroMark Q96 ID pyrosequencer using PyroMark Gold Q96 SQA reagents (all were from Qiagen), or on a Q24 pyrosequencer according to the manufacturer’s protocol (Biotage). The methylation of C in each analysed CpG site was quantified from 0 to 100%, using the PyroMark software (Qiagen).

Table II

Primers for bisulfite pyrosequencing.

Table II

Primers for bisulfite pyrosequencing.

LINE1.2 (6 CpGs)
  LINE-1 fwd: 5′-BIO-TAGGGAGTGTTAGATAGT GG-3′,
  LINE-1 rev: 5′-AACTCCCTAACCCCTTAC-3′,
  LINE-1 seq: 5′-CAAATAAAACAATACCTC-3′.
RASSF1A (9 CpGs)
  RASSF1 methF: 5′-AGTATAGTAAAGTTGGTTTTTAGAAA-3′
  RASSF1 methR: 5′-CCCTTCCTTCCCTCCTT-3′
  RASSF1 methPSEQ: 5′-AAGTTGGTTTTTAGAAATA-3′
VEGFR1 (15 CpGs)
  VEGFR1 PyroF: 5′-AGGAGGGGTAAGGGTAAGAG-3′
  VEGFR1 PyroR: 5′-TCCCCACCTACCCTCTTCTT-3′
  VEGFR1 PyroSEQ: 5′-GGGAGAGGAGTAAAGATTTTGAATT-3′
Western blot analysis

Protein lysates were extracted from cell cultures using RIPA buffer [50 mM Tris HCl, pH 7.4, 150 mM NaCl, 1 mM EDTA, 1% (v/v) Triton X-100, 0.1% (w/v) SDS], supplemented with 10 μl phenylmethylsulfonyl fluoride (87 mg/ml in 96% EtOH) and 100 μl protease inhibitor cocktail (2 mM AEBSF, 1 mM EDTA, 130 μM bestatin, 14 μM E-64, 1 μM leupepin, 0.3 μM aprotinin). Lysates were separated by SDS/PAGE and subsequently transferred onto pre-activated polyvinylidene fluoride nitrocellulose membranes (PVDF). Membranes were blocked for 1 h at room temperature (RT) in TBST [10 mM Tris-HCl (pH 7.5), 10 mM NaCl, 0.1% (v/v) Tween 20] containing 5% nonfat dry milk powder. Membranes were immunoblotted overnight at 4°C in TBST with 5% nonfat dry milk powder with DNMT1 (supplier) with HDAC1 (Cell Signalling Technologies) used as a loading control as appropriate. All secondary antibodies were HRP-labelled and bound antibody complexes were detected using the Supersignal West Pico Chemiluminescent kit (Pierce, Rockford, IL, USA).

Results

Gemcitabine reactives mRNA expression of epigenetically silenced genes

A549 (NSCLC), H28 (MPM), LNCaP (prostate) and 22Rv1 (prostate) cells were treated with either decitabine or gemcitabine and the effects of these drugs on gene expression were examined. In A549 cells decitabine and gemcitabine induced both VEGFR1 and VEGFR2 (Fig. 2A). In the H28 cell line a gene previously shown to be epigenetically silenced in this cell line by DNA CpG methylation (25,26), sFRP4, was reactivated by both drugs (Fig. 2B). Both GSTP1 and IGFBP3 have been shown to be epigenetically silenced by DNA CpG methylation in prostate cancer by us and others (2729). Using quantitative PCR we measured the effect of decitabine and gemcitabine on these genes in two prostate cancer cell lines (LNCaP and 22Rv1). Both drugs were shown to significantly induce expression of GSTP1 (Fig. 2C) and IGFBP3 (Fig. 2D).

As all these genes were examined separately in different cell lines we reviewed the literature to find whether there was a common gene frequently silenced by DNA CpG methylation in lung, MPM and prostate cell lines. From this analysis we chose the gene RASSF1A which was shown to be silenced by methylation in several of our cell lines (3032). Gemcitabine was able to reactivate/upregulate RASSF1A in 3 out of the 4 cell lines tested whereas decitabine reactived RASSF1A in 4 out of the 4 cell lines at all concentrations tested (Fig. 3).

Decitabine functions in vitro and in vivo to inhibit DNA methyltransferases

A DNA methyltransferase activity/inhibition assay was used to measure the effects of gemcitabine on DNMT enzymatic activity, with decitabine used as a positive inhibitor control. Using recombinant DNMT protein mixture we demonstrated that both decitabine and gemcitabine inhibited DNMT activity (Fig. 4A and B). Furthermore, DNMT activity was also inhibited in nuclear extracts from cells exposed to either decitabine or gemcitabine (Fig. 4C and D), indicating that gemcitabine inhibits DNA methyltransferase activity.

Gemcitabine affects DNMT1 protein stability

Decitabine has been shown to selectively degrade DNMT1 protein levels by a proteasomal-based pathway (33). To examine whether gemcitabine also affects the levels of DNMT1 protein, western blot analyses were carried out on extracts from cells treated with various concentrations of gemcitabine or decitabine. Gemcitabine was found to affect DNMT1 protein levels to different degrees for all the cell lines examined (Fig. 4E).

Gemcitabine does not alter global DNA CpG methylation or at the promoters of reactivated genes

Schafer and colleagues published data demonstrating that gemcitabine affects DNA methylation in cancer cells by inhibiting nucleotide excision repair (NER)-mediated DNA demethylation. Intriguingly, the authors showed that gemcitabine treatments did not affect global levels of DNA methylation but induced hypermethylation and silencing of MLH1 (34).

Using pyrosequencing of long interspersed nucleotide element (LINE-1) sequences, we examined global methylation in our cell lines. In agreement with Schafer et al (34), we did not observe any gross alterations in the global DNA CpG methylation following treatment with gemcitabine (Table III). For the lower concentration of decitabine tested (0.2 μM) we noted a decrease in CpG methylation which did not occur in the cells treated at the higher concentration (1 μM) in agreement with effects reported by others (35).

Table III

Percentage of methylation for LINE-1 and RASSF1A as determined by pyrosequencing.

Table III

Percentage of methylation for LINE-1 and RASSF1A as determined by pyrosequencing.

Cell lineTreatmentLINE-1RASSF1
A549UT55.885.2
DAC 0.232.845.3
DAC 1.051.379.2
Gem 0.254.785.8
Gem 1.055.786.2
SKMES-1UT53.17.3
DAC 0.245.24.4
DAC 1.052.76.5
Gem 0.251.25.2
H28UT68.094.9
DAC 0.247.865.8
DAC 1.063.888.1
Gem 0.269.694.6
Gem 1.068.294.5
Du145UT65.271.2
DAC 0.245.851.0
DAC 1.056.564.0
Gem 0.264.371.6
Gem 1.066.071.9
22Rv1UT50.083.8
DAC 0.237.753.3
DAC 1.042.561.2
Gem 0.250.482.6
Gem 1.052.074.3

[i] UT, untreated; DAC 0.2, decitabine 0.2 μM; DAC 1.0, decitabine 1.0 μM; Gem 0.2, gemcitabine 0.2 μM; Gem 1.0, gemcitabine 1.0 μM.

Next, we examined the effect of decitabine and gemcitabine on CpG methylation within RASSF1A. Again decitabine caused alterations to DNA CpG methylation levels at the lower concentration, yet there was no effect on methylation at the higher concentration (Table III). For the most part gemcitabine did not alter DNA CpG methylation for either concentration examined (Table III). Similar results were observed for VEGFR1 (data not shown).

GSTP1 has been previously shown to be downregulated by DNA CpG methylation (2729). Given that we were able to demonstrate that both gemcitabine and decitabine reactivated the expression of GSTP1 in prostate cancer cell lines (Fig. 2), using MS-PCR, we examined whether or not there is a loss of methylation at the GSTP1 promoter in the 22Rv1 cell line. Our results showed a clear increase in the levels of unmethylated DNA at the GSTP1 promoter following treatment with either drug, indicating that there may be some changes to DNA CpG methylation occurring in cells treated with gemcitabine (Fig. 5).

Discussion

Gemcitabine is a well-established chemotherapy agent used as a single-agent treatment in pancreatic cancer, or in combination with various other agents in the treatment of many solid tumours. Its mode of action is generally considered to be that of an anti-metabolite acting to either inhibit ribonucleotide reductase (RNR) or by replacing cytidine during DNA replication.

Aberrant epigenetic regulation through DNA CpG methylation is a frequent event in cancer and reactivation of the target of methylation induced silencing 1 (TMS1) by DNA methyltransferase inhibitors (DNMTi) was found to enhance sensitivity to gemcitabine in pancreatic cancer cells (36).

We noted that gemcitabine shares structural similarity with the FDA-approved DNMTi decitabine (Dacogen) (Fig. 1) and tested whether or not it has the ability to affect DNA CpG methylation. We showed that gemcitabine actively induces expression of epigenetically silenced genes in lung, mesothelioma (MPM) and prostate cancer cell lines (Figs. 2 and 3). We showed that gemcitabine directly inhibits DNA methyltransferases (Fig. 4). In addition, we also showed that gemcitabine depletes levels of DNMT1 similar to decitabine (Fig. 4). However, despite gemcitabine’s ability to inhibit DNMTs, alter cellular DNMT1 protein levels and reactivate epigenetically silenced genes, there were few if any changes to DNA CpG methylation levels. We provide some evidence that active demethylation of an epigenetically silenced gene can occur (Fig. 5), but another mechanism may have to be invoked.

Schafer et al (34) published additional data demonstrating that gemcitabine affects DNA methylation in cancer cells by inhibiting nucleotide excision repair (NER)-mediated DNA demethylation. The authors showed that gemcitabine treatments did not affect global levels of DNA methylation but induced hypermethylation and silencing of MLH1 (34). In agreement with the authors we also found that global levels of DNA CpG methylation were unaffected. In addition, methylation at the promoters of genes which could be reactivated following treatments with either decitabine or gemcitabine did not demonstrate any major changes in methylation status. This may in part be due to the concentrations of drugs used. For example, it has been established that high concentrations of decitabine have less effect on DNA methylation due to cytostatic effects, and our results using pyrosequencing confirm this demonstrating that changes in DNA methylation only occurred at the lower concentration of drug used (Table III). Using MTT assays we examined cellular proliferation across a range of concentrations (decitabine 200–5,000 nM; gemcitabine 50–1,000 nM) in A549 and SKMES1 cells. While decitabine did not affect cellular proliferation to any appreciable effect at any concentration, gemcitabine caused significant decreases (of the order 25–50%) in cellular proliferation (data not shown), which was not unexpected given the other known mechanisms of action of this drug.

Schafer et al (34) used a range of concentrations (34, 67 or 134 nM), whereas we used higher concentrations (0.2–1 μM). It must be noted, however, that in gemcitabine infusion studies of patients with solid tumours, blood concentrations of gemcitabine at Day 1 of infusion varied between 18 and 77 μM and at Day 15 between 13 and 90 μM (37). As such the amount of gemcitabine used in our analysis was still below the levels utilised in patients.

The concentration of gemcitabine may become a critical determinant in how this chemotherapeutic agent is used in the clinic. For instance, in a study using MPM cells, gemcitabine inhibited the secretion of the proinflammatory cytokine IL-6 at drug concentrations that produced only small decreases in cell viability, whereas at higher doses a surge of IL-6 release was noted (38). We also observed an increase in the protumourigenic cytokine IL-23 in a NSCLC cell line treated with gemcitabine (Gray and O’Byrne, unpublished data).

How then does gemcitabine induce or reactivate epigenetically silenced genes. The nitrogen substitution of carbon 5 in the cytosine ring (Fig. 1) is involved in the mechanism of DNMT inactivation. As this nitrogen is not present in gemcitabine it may be that some sort of steric hindrance is involved affecting DNMTs and or methyl-binding proteins. Conceivably by inhibiting binding of methyl-binding protein complexes to DNA CpG sites this may relax chromatin and result in the re-expression of silenced genes.

The results presented by us and Schafer et al (34) demonstrate a novel indication for gemcitabine and may have important implications in oncology as DNA methylation effects are potentially useful as biomarkers to either monitor response to therapy (39), or may have predictive value (40). In particular, the ability of gemcitabine to reactivate epigenetically silenced genes indicates a potential means to monitor and identify those patients who actually respond to gemcitabine treatment.

There are exciting new avenues which could be exploited, for example the use of low dose gemcitabine regimens combined with other epigenetic targeting drugs such histone deacetylase inhibitors. In addition low dose regimens may have the ability to sensitise resistant cancer to other cancer therapies. Further study will be required to fully delineate these possibilities.

Acknowledgements

Funding support for this project was provided by the Irish Cancer Society to Antoinette Perry.

References

1. 

M HidalgoPancreatic cancerN Engl J Med36216051617201010.1056/NEJMra0901557

2. 

VT DeVita JrA step in the right directionNat Clin Pract Oncol3641200610.1038/ncponc066917139311

3. 

KH TkaczukReview of the contemporary cytotoxic and biologic combinations available for the treatment of metastatic breast cancerClin Ther3122732289200910.1016/j.clinthera.2009.11.01120110041

4. 

J GoffinC LacchettiPM EllisYC UngWK EvansFirst-line systemic chemotherapy in the treatment of advanced non-small cell lung cancer: a systematic reviewJ Thorac Oncol5260274201010.1097/JTO.0b013e3181c6f035

5. 

S NagelR CalifanoN ThatcherF BlackhallGemcitabine and carboplatin in combination for the treatment of advanced, metastatic, non-small cell lung cancerExpert Opin Pharmacother832653275200710.1517/14656566.8.18.326518035969

6. 

DA FennellG GaudinoKJ O’ByrneL MuttiJ van MeerbeeckAdvances in the systemic therapy of malignant pleural mesotheliomaNat Clin Pract Oncol5136147200810.1038/ncponc103918227828

7. 

M RayHL KindlerMalignant pleural mesothelioma: an update on biomarkers and treatmentChest136888896200910.1378/chest.08-266519736192

8. 

P JantscheffN EsserR GraeserLiposomal gemcitabine (GemLip)-efficient drug against hormone-refractory Du145 and PC-3 prostate cancer xenograftsProstate6911511163200910.1002/pros.2096419399788

9. 

P JantscheffV ZiroliN EsserAnti-metastatic effects of liposomal gemcitabine in a human orthotopic LNCaP prostate cancer xenograft modelClin Exp Metastasis26981992200910.1007/s10585-009-9288-119784785

10. 

W PlunkettP HuangYZ XuV HeinemannR GrunewaldV GandhiGemcitabine: metabolism, mechanisms of action, and self-potentiationSemin Oncol22Suppl 1131019957481842

11. 

MW LawlessS NorrisKJ O’ByrneSG GrayTargeting histone deacetylases for the treatment of diseaseJ Cell Mol Med13826852200910.1111/j.1582-4934.2008.00571.x19175682

12. 

KJ O’ByrneMP BarrSG GrayThe role of epigenetics in resistance to cisplatin chemotherapy in lung cancerCancers314261453201124212667

13. 

D CaiDS ShamesMG RasoSteroid receptor coactivator-3 expression in lung cancer and its role in the regulation of cancer cell survival and proliferationCancer Res7064776485201010.1158/0008-5472.CAN-10-000520663904

14. 

N HajjiK WallenborgP VlachosJ FullgrabeO HermansonB JosephOpposing effects of hMOF and SIRT1 on H4K16 acetylation and the sensitivity to the topoisomerase II inhibitor etoposideOncogene2921922204201010.1038/onc.2009.50520118981

15. 

W ZhaoQ ZhangX KangS JinC LouAIB1 is required for the acquisition of epithelial growth factor receptor-mediated tamoxifen resistance in breast cancer cellsBiochem Biophys Res Commun380699704200910.1016/j.bbrc.2009.01.15519285025

16. 

SB BaylinPA JonesA decade of exploring the cancer epigenome - biological and translational implicationsNat Rev Cancer11726734201110.1038/nrc313021941284

17. 

SD GoreC JonesP KirkpatrickDecitabineNat Rev Drug Discov5891892200610.1038/nrd218017117522

18. 

E KaminskasA FarrellS AbrahamApproval summary: azacitidine for treatment of myelodysplastic syndrome subtypesClin Cancer Res1136043608200510.1158/1078-0432.CCR-04-213515897554

19. 

T YokochiKD RobertsonDoxorubicin inhibits DNMT1, resulting in conditional apoptosisMol Pharmacol6614151420200410.1124/mol.104.00263415340041

20. 

SG GrayN Al-SarrafAM BairdMC CathcartE McGovernKJ O’ByrneRegulation of EP receptors in non-small cell lung cancer by epigenetic modificationsEur J Cancer4530873097200910.1016/j.ejca.2009.09.00619818596

21. 

PW LairdA ZijderveldK LindersMA RudnickiR JaenischA BernsSimplified mammalian DNA isolation procedureNucleic Acids Res194293199110.1093/nar/19.15.42931870982

22. 

A DaskalosS LogothetiS MarkopoulouGlobal DNA hypomethylation-induced DeltaNp73 transcriptional activation in non-small cell lung cancerCancer Lett3007986201110.1016/j.canlet.2010.09.00920926182

23. 

A DaskalosG NikolaidisG XinarianosHypomethylation of retrotransposable elements correlates with genomic instability in non-small cell lung cancerInt J Cancer1248187200910.1002/ijc.2384918823011

24. 

A DaskalosU OleksiewiczA FiliaUHRF1-mediated tumor suppressor gene inactivation in nonsmall cell lung cancerCancer11710271037201110.1002/cncr.2553121351083

25. 

B HeAY LeeS DadfarmaySecreted frizzled-related protein 4 is silenced by hypermethylation and induces apoptosis in beta-catenin-deficient human mesothelioma cellsCancer Res65743748200515705870

26. 

AY LeeB HeL YouExpression of the secreted frizzled-related protein gene family is downregulated in human mesotheliomaOncogene2366726676200410.1038/sj.onc.120788115221014

27. 

WG NelsonAM De MarzoS YegnasubramanianEpigenetic alterations in human prostate cancersEndocrinology15039914002200910.1210/en.2009-057319520778

28. 

AS PerryH LiyanageM LawlerK WoodsonDiscovery of DNA hypermethylation using a DHPLC screening strategyEpigenetics24349200710.4161/epi.2.1.388217965617

29. 

AS PerryB LoftusR MorooseIn silico mining identifies IGFBP3 as a novel target of methylation in prostate cancerBr J Cancer9615871594200710.1038/sj.bjc.660376717453001

30. 

H EndohY YatabeS ShimizuRASSF1A gene inactivation in non-small cell lung cancer and its clinical implicationInt J Cancer1064551200310.1002/ijc.1118412794755

31. 

I KuzminJW GillespieA ProtopopovThe RASSF1A tumor suppressor gene is inactivated in prostate tumors and suppresses growth of prostate carcinoma cellsCancer Res6234983502200212067994

32. 

S ToyookaHI PassN ShivapurkarAberrant methylation and simian virus 40 tag sequences in malignant mesotheliomaCancer Res6157275730200111479207

33. 

K GhoshalJ DattaS Majumder5-Aza-deoxycytidine induces selective degradation of DNA methyltransferase 1 by a proteasomal pathway that requires the KEN box, bromo-adjacent homology domain, and nuclear localization signalMol Cell Biol2547274741200510.1128/MCB.25.11.4727-4741.2005

34. 

A SchaferL SchomacherG BarretoG DoderleinC NiehrsGemcitabine functions epigenetically by inhibiting repair mediated DNA demethylationPLoS One5e14060201010.1371/journal.pone.001406021124914

35. 

HC TsaiH LiL Van NesteTransient low doses of DNA-demethylating agents exert durable antitumor effects on hematological and epithelial tumor cellsCancer Cell21430446201210.1016/j.ccr.2011.12.02922439938

36. 

K RamachandranH MillerE GordianC Rocha-LimaR SingalMethylation-mediated silencing of TMS1 in pancreatic cancer and its potential contribution to chemosensitivityAnticancer Res3039193925201021036703

37. 

SM de LangeK van der BornJR KroepNo evidence of gemcitabine accumulation during weekly administrationEur J Clin Pharmacol61843849200516283278

38. 

BR McLarenBW RobinsonRA LakeNew chemotherapeutics in malignant mesothelioma: effects on cell growth and IL-6 productionCancer Chemother Pharmacol45502508200010.1007/s00280005102610854139

39. 

VV LevensonDNA methylation as a universal biomarkerExpert Rev Mol Diagn10481488201010.1586/erm.10.1720465502

40. 

F SalazarMA MolinaM Sanchez-RoncoFirst-line therapy and methylation status of CHFR in serum influence outcome to chemotherapy versus EGFR tyrosine kinase inhibitors as second-line therapy in stage IV non-small-cell lung cancer patientsLung Cancer728491201110.1016/j.lungcan.2010.07.00820705357

Related Articles

Journal Cover

December 2012
Volume 30 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gray SG, Baird A, O'Kelly F, Nikolaidis G, Almgren M, Meunier A, Dockry E, Hollywood D, Ekström TJ, Perry AS, Perry AS, et al: Gemcitabine reactivates epigenetically silenced genes and functions as a DNA methyltransferase inhibitor. Int J Mol Med 30: 1505-1511, 2012
APA
Gray, S.G., Baird, A., O'Kelly, F., Nikolaidis, G., Almgren, M., Meunier, A. ... O'Byrne, K. . (2012). Gemcitabine reactivates epigenetically silenced genes and functions as a DNA methyltransferase inhibitor. International Journal of Molecular Medicine, 30, 1505-1511. https://doi.org/10.3892/ijmm.2012.1138
MLA
Gray, S. G., Baird, A., O'Kelly, F., Nikolaidis, G., Almgren, M., Meunier, A., Dockry, E., Hollywood, D., Ekström, T. J., Perry, A. S., O'Byrne, K. ."Gemcitabine reactivates epigenetically silenced genes and functions as a DNA methyltransferase inhibitor". International Journal of Molecular Medicine 30.6 (2012): 1505-1511.
Chicago
Gray, S. G., Baird, A., O'Kelly, F., Nikolaidis, G., Almgren, M., Meunier, A., Dockry, E., Hollywood, D., Ekström, T. J., Perry, A. S., O'Byrne, K. ."Gemcitabine reactivates epigenetically silenced genes and functions as a DNA methyltransferase inhibitor". International Journal of Molecular Medicine 30, no. 6 (2012): 1505-1511. https://doi.org/10.3892/ijmm.2012.1138